WO1995034573A1 - Identification of polycystic kidney disease gene, diagnostics and treatment - Google Patents

Identification of polycystic kidney disease gene, diagnostics and treatment Download PDF

Info

Publication number
WO1995034573A1
WO1995034573A1 PCT/US1995/007079 US9507079W WO9534573A1 WO 1995034573 A1 WO1995034573 A1 WO 1995034573A1 US 9507079 W US9507079 W US 9507079W WO 9534573 A1 WO9534573 A1 WO 9534573A1
Authority
WO
WIPO (PCT)
Prior art keywords
pkd1
gene product
pkd1 gene
sep
gene
Prior art date
Application number
PCT/US1995/007079
Other languages
French (fr)
Other versions
WO1995034573A9 (en
Inventor
Stephen Reeders
Michael Schneider
Sandra Glucksmann
Original Assignee
Brigham And Women's Hospital
Millenium Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Brigham And Women's Hospital, Millenium Pharmaceuticals, Inc. filed Critical Brigham And Women's Hospital
Priority to AU27661/95A priority Critical patent/AU2766195A/en
Publication of WO1995034573A1 publication Critical patent/WO1995034573A1/en
Publication of WO1995034573A9 publication Critical patent/WO1995034573A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Abstract

The present invention relates to the identification of the autosomal dominant polycystic kidney disease (PKD) gene and high throughput assays to identify compounds that interfere with PKD activity. Interfering compounds that inhibit the expression, synthesis and/or bioactivity of the PKD gene product can be used therapeutically to treat polycystic kidney disease.

Description


  
 



   IDENTIFICATION OF POLYCYSTIC KIDNEY
 DISEASE GENE, DIAGNOSTICS AND TREATMENT
 This is a continuation-in-part of U.S. Serial No.



  08/253,524, filed, June 3, 1994, which is incorporated by reference herein in its entirety.



   1. INTRODUCTION
 The present invention relates to the identification of the gene, referred to as the PKD1 gene, mutations in which are responsible for the vast majority of cases involving autosomal dominant polycystic kidney disease (ADPKD). The
PKD1 gene, including the complete nucleotide sequence of the gene's coding region are presented. Further, the complete
PKD1 gene product amino acid sequence and protein structure and antibodies directed against the PKD1 gene product are also presented. Additionally, the present invention relates to therapeutic methods and compositions for the treatment of
ADPKD symptoms. Methods are also presented for the identification of compounds that modulate the level of expression of the PKD1 gene or the activity of mutant PKD1 gene product, and the evaluation and use of such compounds in the treatment of ADPKD symptoms.

  Still further, the present invention relates to prognostic and diagnostic, including prenatal, methods and compositions for the detection of mutant PKD1 alleles and/or abnormal levels of PKD1 gene product or gene product activity.



   2. BACKGROUND OF THE INVENTION
 Autosomal dominant polycystic kidney disease (ADPKD) is among the most prevalent dominant human disorders, affecting between 1 in 1,000 and 1 in 3,000 individuals worldwide (Dalgaard, O.Z., 1957, Acta. Med. Scand. 158:1-251). The major manifestation of the disorder is the progressive cystic dilation of renal tubules (Gabow, P.A., 1990, Am. J. Kidney
Dis. 16:403-413), leading to renal failure in half of affected individuals by age 50.  



   ADPKD-associated renal cysts may enlarge to contain several liters of fluid and the kidneys usually enlarge progressively causing pain. Other abnormalities such as pain, hematuria, renal and urinary infection, renal tumors, salt and water imbalance and hypertension frequently result from the renal defect. Cystic abnormalities in other organs, including the liver, pancreas, spleen and ovaries are commonly found in ADPKD. Massive liver enlargement occasionally causes portal hypertension and hepatic failure.



  Cardiac valve abnormalities and an increased frequency of subarachnoid and other intracranial hemorrhage have also been observed in ADPKD. Progressive renal failure causes death in many ADPKD patients and dialysis and transplantation are frequently required to maintain life in these patients.



  Although end-stage renal failure usually supervenes in middle age (ADPKD is sometimes called adult polycystic kidney disease), children may occasionally have severe renal cystic disease.



   Although studies of kidneys from ADPKD patients have demonstrated a number of different biochemical, structural and physiological abnormalities, the disorder's underlying causative biochemical defect remains unknown. Biochemical abnormalities which have been observed have involved proteinsorting, the distribution of cell membrane markers within renal epithelial cells, extracellular matrix, ion transport, epithelial cell turnover, and epithelial cell proliferation.



  The most carefully documented of these findings are abnormalities in the composition of tubular epithelial cells, and a reversal of the normal polarized distribution of cell membrane proteins, such as the   Na+/K    ATPase (Carone, F.A. et al., 1994, Lab. Inv. 70:437-448.).



   As the name implies, ADPKD is inherited as an autosomal dominant disorder. Three distinct loci have been shown to cause phenotypically indistinct forms of the disease, with greater than   85-90W    of disease incidence being due to mutations which map to the short arm of chromosome 16, as  discussed below. Despite intensive investigation, the molecular defect responsible for ADPKD is not known.



   In 1985 Reeders et al. (Reeders et al., Nature 317:542, 1985) carried out genetic linkage studies of a large number of ADPKD families and demonstrated that a gene on the short arm of chromosome 16 was mutated in most cases of ADPKD.



  This gene has been designated PKD1 by the Nomenclature
Committee of the Human Gene Mapping Workshop and the Genome
Data Base of the Welch library, John Hopkins University.



  Further linkage studies have identified a set of genetic markers that flank the gene-rich region containing the PKD1 gene (Reeders et al.,   1988,    Genomics 3:150; Somlo et al., 1992, Genomics 13:152; Breuning et al., 1990, J. Med. Genet.



  27:603; Germino et al., 1990, Am. J. Hum. Genet. 46:925).



  These markers have been mapped by a variety of physical mapping techniques including fluorescent in situ hybridization and pulsed-field gel electrophoresis (Gillespie et al., 1990, Nucleic Acids Research 18:7071). It has been shown that the closest distal genetic marker (D16S259; on the telomeric side of the PKD1 locus) lies within 750 kb of the closest proximal genetic marker (D16S25; on the centromeric side of the PKD1 locus). The interval between the genetic markers has been cloned in a series of overlapping cosmid and bacteriophage genomic clones (Germino et al., 1992, Genomics 13:144), which contain the entire PKD1 interval, with the exception of two gaps of less than 10 kb and less than 50 kb.



  Restriction mapping of these clones has confirmed that the interval between the flanking genetic markers is 750 kb.



   While genetic mapping studies such as these have begun to narrow the region within the human genome in which the gene responsible for ADPKD lies, there exist an estimated twenty or more genes within this 750 kb interval. Given the prevalence and severity of ADPKD, however, it is of great importance to eludicate which, if any, of these postulated genes corresponds to PKD1.  



   3. SUMMARY OF THE INVENTION
 The present invention relates to methods and compositions for the diagnosis and treatment of autosomal dominant polycystic kidney disease (ADPKD). Specifically, a novel gene, referred to as the PKD1 gene, is described in
Section 5.1. Mutations within the PKD1 gene are responsible for approximately 90% cases of ADPKD. Additionally, the PKD1 gene product, including the nucleotide sequence of the complete coding region is described in Section 5.2.



  Antibodies directed against the PKD1 gene product are described in Section 5.3.



   Further, the present invention relates to therapeutic methods and compositions for the amelioration of ADPKD symptoms. These therapeutic techniques are described in
Sections 5.9 and 5.10. Methods are additionally presented for the identification of compounds that modulate the level of expression of the PKD1 gene or the activity of PKD1 mutant gene products, and the evaluation and use of such compounds as therapeutic ADPKD treatments. Such methods are described in Section 5.8.



   Still further, the present invention relates to prognostic and diagnostic, including prenatal, methods and compositions whereby the PKD1 gene and/or gene product can be used to identify individuals carrying mutant   PKDl    alleles, exhibiting an abnormal level of PKD1 gene product or gene product activity. Additionally, the present invention describes methods which diagnose subjects exhibiting ADPKD symptoms. Such techniques are described in Section 5.12.

 

  Additionally, the present invention relating to the use of
PKD1 animal knockout screening assays for the identification of compounds useful for the amelioration of ADPKD symptoms.



   The coding region of the PKD1 gene is complex and extensive, having a size of approximately 60 kb and containing a total of 46 exons, the sequence of which, until now, has been difficult to obtain for a number of reasons.



  First, the majority (approximately the first two thirds) of the PKD1 gene is duplicated several times in a transcribed  fashion elsewhere in the genome, thus making it very difficult to distinguish authentic PKD1 sequence from PKD1like sequence. Further, the PKD1 gene contains extensive repeated regions of high GC content which are not only difficult to sequence accurately, but, additionally, make the alignment of PKD1 nucleotide sequence extremely difficult.



  Still further, the PKD1 gene encodes a large transcript of approximately 14.5 kb in length, and evidence exists that there are alternatively spliced forms of the gene. Thus, the size of the PKD1 gene, the size and complexity of PKD1 transcript, coupled with the above-described PKD1 features made the successful sequencing of the gene and its cDNA very difficult. As described in Sections 5.1.2 and in the Example presented in Section 10, below, however, the obstacles to sequencing the PKD1 gene have now, for the first time, been overcome.



   The PKD1 transcript, which is approximately 14.5 kb in length, encodes a PKD1 gene product with a derived amino acid sequence of 4304 amino acid residues. This PKD1 gene product contains at least five distinct peptide domains which are likely to be involved in protein-protein and/or protein carbohydrate interactions. Further, this PKD1 gene product shares amino acid sequence similarity with a number of extracellular matrix proteins. These features of the PKD1 gene product indicate that ADPKD is caused by a biochemical defect involving extracellular signalling and/or extracellular matrix assembly, and suggests therapeutic strategies whereby ADPKD can be treated and/or whereby ADPKD symptoms can be ameliorated.



   The Examples described in Section 6 through 11, below, demonstrate the successful identification and characterization of the PKD1 gene and gene product, including the complete nucleotide sequence of the PKD1 coding region, the complete amino acid sequence, and the elucidation of the protein structure of the PKD1 gene product. Further, a
ADPKD-causing mutation is identified and described.  



   4. DESCRIPTION OF THE FIGURES
 FIG. 1. A map of the PKD1 interval showing the cosmids and bacteriophage clones covering the region (Taken from
Germino et al, 1992, Genomics 13:144.) The PKDI region as defined by flanking markers extends from D16S259   (pGGG1)    to
D16S25, a span of approximately 750kb. Single-copy probes used in pulsed-field gel mapping of the region are shown above the line (pGGG1, CMM65b, etc.). C, M, P, N and B are sites for restriction enzymes ClaI,   MluI,    PvuI, NotI and
BssHII, respectively. Sites that cleave in genomic DNA from only some tissues are shown in parenthesis. Bold bars (a-z, aa) represent the extents of the coding regions (see Table 2).

  Horizontal lines 1-38 represent cosmid and phage clones spanning the PKD1 region, as shown here:   l=cJC1    9=cDEB11 17=cKLH4 25=cNK30 2=cJC2   10=cGGG10    18=cKLH6   26=ALCNlwl      3=cDEBl      11=cGGGl    19=cKLH7   27=ALCNw2J2    4=CDEB4 12=cGGG2 20=cKLH8 28=XLCNwlw3 5-cDEB7 13-cGGG3 21=cKLH9   29=ALCNw5.2      6=cDEB8    14-cGGG4a 22-cNK32 30=XNK92.6w5.1 7=cDEB9 15=cGGG4b   23=cNK31      31=ANK92.6w4.1      8=cDEB10    16=cGGG6 24=cGGG8 32=cNK92.6w1.3 33=cNK92.6    34=cNK92 .2 35=cNK63 .7 36=cNK14    37=cCOS4 38=cCOS3
 FIG. 2. A map of the PKD1 region as defined by flanking markers.

  The region extends from D16S259 (pGGGl) to w5.2CA, a microsatellite repeat that lies within ALCNw5.2, a span of approximately 480kb. The labels are as for FIG. 1.



   FIG. 3A-B. Genomic DNA from 40 unrelated ADPKD patients was amplified by PCR for SSCP analysis. Primers F23 and R23 (See Table 1, below) were used to amplify an exon of 298bp.



  Variant SSCP patterns were seen in two ADPKD patients under the following conditions. Each of the patients was heterozygous for the normal pattern and the variant pattern.  



  The pattern seen in these patients was not seen in normal individuals. Arrow indicates non-denatured DNA.



   FIG. 4. A map (not to scale), derived from the cosmid contig cGGG1, cGGG10 and cDEB11, of the genomic region containing the PKD1 gene. The horizontal black bars show the positions of the three cosmids. The discontinuities in these bars indicate that the full extent of cGGG1 and cDEB11 are not shown. The map was constructed using restriction enzyme data from several enzymes. BamHI, EcoRI and NotI restriction sites are shown. The numbers below the horizontal line represent distances in kilobases between adjacent restriction sites. The PKD1 cDNA clones are shown above as grey bars.



  These clones hybridize to the restriction fragments shown immediately below them in the genomic map.



   FIG. 5A. Structure of the PKD1 gene transcript. The bar at the top represents the PKD1 exon map. A total of 46 exons were identified. Below the gene transcript map are depictions of the overlapping cDNA clones, with putative alternatively spliced regions as indicated.



   FIGS.   5B-SC.    PKD1 exons. This chart lists PKD1 exon sizes and indicates which cDNA clones contain nucleotide sequences corresponding to sequences present within specific exons.



   FIG. 6. PKD1 nucleotide and amino acid sequences.



  Depicted herein are, top line, the nucleotide sequence of the entire PKD1 coding region (SEQ ID NO: 1), and, bottom line, the PKD1 derived amino acid sequence (SEQ ID NO: 2), given in the one-letter amino acid code.



   FIGS. 7A-7B. The derived amino acid sequence of PKD1 gene product (SEQ ID NO: 2). The putative peptide domains of the
PKD1 gene product are depicted underneath the amino acid sequence.  



   FIG. 8. A schematic representation of the PKD1 gene product, with each of its putative domains illustrated.



   FIG. 9. SSCP analysis. Genomic DNA from a total of 60 unrelated ADPKD patients was amplified by PCR for SSCP analysis. Intronic primers F25 and   Mill-lR    (see Section 10.1, below) were used for amplification. A variant SSCP pattern was seen in one individual. The amplified DNA from this individual was then reamplified with the intronic primers KG8-F31 and KG8-R35 (see Section 10.1, below). Both strands of the reamplied DNA were sequenced, using F25 and   Mill-lR    as sequencing primers. As discussed in Section 10.2, below, sequencing revealed a C to T transition which created a stop codon at PKD1 amino acid position 765. The pattern seen in these patients was not seen in normal individuals.



   5. DETAILED DESCRIPTION OF THE INVENTION
 Methods and compositions for the diagnosis and treatment of (ADPKD) are described herein. Specifically, the gene, referred to herein as the PKD1 gene, in which mutations occur that are responsible for the vast majority of ADPKD cases is described. Further, the PKD1 gene product and antibodies directed against the PKD1 gene product are also presented.

 

   Therapeutic methods and compositions are described for the treatment and amelioration of ADPKD symptoms. Further, methods for the identification of compounds that modulate the level of expression of the PKD1 gene or the activity of mutant PKD1 gene product, and the evaluation and use of such compounds in the treatment of ADPKD symptoms are also provided.



   Still further, prognostic and diagnostic methods are described for the detection of mutant PKD1 alleles, of abnormal levels of PKD1 gene product or of gene product activity.  



   5.1. THE PKD1 GENE
 The PKD1 gene, mutations in which are responsible for greater than 9 in 10 cases of ADPKD, is described herein.



  Specifically, the strategy followed to identify the PKD1 gene is briefly discussed, as is the strategy for obtaining the complete nucleotide sequence of the gene. Further, the PKD1 nucleotide sequence and alternative splicing features are described. Still further, nucleic acid sequences that hybridize to the PKD1 gene and which may be utilized as therapeutic ADPKD treatments and/or as part of diagnostic methods are described. Additionally, methods for the production or isolation of such PKD1 nucleic acid molecules and PKD1-hybridizing molecules are described.



     5.1.1. IDENTIFICATION    OF THE PKD1 GENE
 Prior to the present invention, it had only been known that the physical location of the PKD1 gene within the human genome was somewhere within a 750 kb chromosomal region on the short arm of chromosome 16. As presented herein, the interval in which this gene lies has now been reduced until the specific PKD1 gene has been identified out of this large portion of DNA.



   Briefly, the strategy which was followed to identify the
PKD1 gene is as described herein. First, as demonstrated in the Example presented in Section 6, below, the 750 kb PKD1 interval was first substantially narrowed to approximately 460 kb, via genetic linkage studies. Next, as shown in the
Example presented in Section 7, below, a maximum of 27 transcriptional units (TUs) were identified within this approximately. 460 kb PKD1 interval. The total length of these TUs was approximately 300 kb. Thus, the region containing the PKD1 coding region was narrowed down to a region of approximately 300 kb.



   Next, as presented in the Example shown in Section 9, below, a Northern analysis was conducted with mRNA isolated from normal and ADPKD patient kidney tissue, in order to attempt to compare the pattern of ADPKD pathology to the  expression profile of the TUs within the PKD1 interval. One of the TUs, Nik9, was eliminated by such an analysis, which indicated undetectable expression in the kidney and liver.



   In addition, as demonstrated in the Example presented in
Section 9, below, a systematic search was undertaken using several independent techniques, including Southern analysis
SSCP, DGGE and direct sequencing of coding sequences, to detect mutations in ADPKD patients within the TUs of the PKD1 region. By conducting such a mutation screen, greater than 80% of the combined identified coding sequences in the PKD1 region were excluded, thus further substantially narrowing down the region in which the PKD1 gene could lie. The screen was initially performed on individual genes until virtually all the coding sequences were shown to be devoid of mutations. The focus on possible PKD1 candidates was further honed by the recognition that PKD1 demonstrated one of the highest new mutation rates known for human diseases.

  Based on this observation, it was hypothesized that either the PKD1 gene contained a highly mutable site or that the gene presented a large number of potential mutation sites, each mutable at a regular frequency. Such a hypothesis is supported by the absence of substantial linkage disequilibrium among selected population groups. Further, this hypothesis predicted that if the PKD1 gene was a small transcript, it should contain a highly mutable element.



   Trinucleotide repeat expansion represent one of the major sources for dominant mutations such as the ADPKD-causing mutations which arise in the PKD1 gene. A systematic search for such highly mutable trinucleotide repeats was conducted within the TUs in the remaining region wherein PKD1 could lie, but no such repeats were identified.



   The only other explanation for the high mutational prevalence is that the gene is physically large and presents a large target for mutations. Of the TUs, nik823, within the potential PKD1 region that had not been excluded by other means, only two were of a size that could potentially support such a high mutation rate. As demonstrated in the Example  presented, below, in Section 9, a search for ADPKD correlative mutations within one of these TUs failed to identify any such mutations, causing it to be excluded as a candidate PKD1 gene. Ultimately, as demonstrated in the
Example presented in Section 10, below, one of these polymorphisms has been shown to be a de novo mutation which is predicted to lead to the production of a truncated PKD1 protein in the affected individual, These finding are highly suggestive, if not proof, that the identified gene is the
PKD1 gene.



   Thus, the examples presented below in Sections 6 through 11 demonstrate, through a variety of techniques, the genetic and molecular characterization of the PKD1 region, and ultimately demonstrate that the PKD1 gene, dominant mutations in which cause ADPKD, has been identified.



   5.1.2.   SEOUENCING    OF THE PKD1 GENE
 As discussed, below, in Section 5.1.3, the nucleotide sequence of the entire coding region of the PKD1 gene has now successfully been isolated and sequenced. In order to achieve this goal, however, a number of PKD1-specific impediments had to be overcome. The strategy for obtaining the PKD1 gene sequence is discussed, briefly, in this
Section. The Example presented below, in Section 11, discusses this sequencing strategy in more detail.



   First, the PKD1 gene is very large, (approximately 60 kb), as is the PKD1 transcript, being approximately 14.5 kb in length. In addition to this size difficulty, approximately two thirds of the 5' end of the gene is duplicated several times in a highly similar, transcribed fashion elsewhere in the human genome (Germino, G.G. et al., 1992, Genomics   13:144-151;    European Chromosome 16 Tuberous
Sclerosis Consortium, 1993, Cell 75:1305-1315).



   The near-identity of the sequence of cDNA derived from
PKD1 and from the PKDl-like duplications made the likelihood of piecing together a full-length PKD1 transcript by merely screening cDNA libraries via hybridization very low. Such a  screening method would be as likely to identify transcripts originating from both the PKDl-like duplicated regions as from the authentic PKD1 locus. In fact, if each of the duplicated loci were as transcriptionally active as the auhentic PKD1 locus, the representation of authentic PKD1 cDNA clones among the total positive clones, would be very low.



   Thus, a strategy was developed for obtaining the authentic   PKDl    sequence which included, first, a plan for obtaining the highest quality of both genomic sequence spanning the duplicated region as well as obtaining duplicate coverage of cDNA sequence spanning the expected length of the
PKD1 transcript; second, to compare the cDNA sequences to the genomic sequence spanning the duplicated region, thus identiying PKD1 exons; and, finally, to assemble the identified exons into a full-length PKD1 coding sequence.



  The isolation of both PKD1 genomic and cDNA sequence and, further, the aligning of such sequences, however, proved to be very difficult.



   PKD1 genomic DNA (whch totals approximately 60 kb) proved to be particularly difficult to characterize for a number of reasons. First, portions of PKD1 genomic DNA (specifically, regions within cosmid   cGGG10)    tended to be preferentially subcloned. For example, screens for trinucleotide repeats in the cGGG10 cosmid identified one
CCT-positive subclone in a Sau3A-generated library of cGGG10 sublcones. This region was, however, vastly underrepresented in both the Sau3A library (i.e., approximately 1 clone out of over 10,000) and subsequent sheared cosmid libraries (in which no such clones were isolated). A plasmid sublone containing the region, G13, proved difficult to grow and to sequence. Sequence analysis of the clone revealed a highly monotonous series of purines (A and G). 

  Such sequences are thought to make the clone difficult to stably propagate in bacteria. Thus, in order to ascertain the level of representation of the cosmid, it was necessary to construct a detailed physical map of the cGGG10 cosmid.  



   Second, genomic sequence within the PKDl region is very
GC-rich (approximately 70%), and forms extensive, stable secondary structures. These PKD1 genomic DNA features made the task of obtaining accurate nucleotide sequence very difficult. Several alternative sequencing conditions, including different polymerases, melting conditions, polymerization conditons and combinations thereof had to be utilized before such sequence was obtained. However, even when reliable nucleotide sequence became available, the extensive amount of repeated sequences within the genomic made the aligning of sequence information very difficult. It became necesary for accurate aligning of sequences, therefore, to use the fine physical map which had been created earlier.



   The sequencing of PKD1 cDNA also presented a number of
PKD1-specific difficulties. First, the 14 kb size of the transcript made it impossible to isolate a single cDNA clone containg the entire PKD1 transcript. Overlapping partial cDNA clones, therefore, had to be obtained in order to piece together an entire sequence. Partial cDNA clones were obtained by sequencing the ends of one cDNA insert, synthesizing probes using this sequence, and obtaining overlapping cDNA clones by their hybridization to such probes. Second, the PKD1 gene was poorly represented in renal cDNA libraries, and, in fact, its expression appeared to be low in a number of tissues, making the isolation of
PKD1 cDNA clones especially difficult.



   5.1.3. THE PKD1 GENE
 Described, herein is the complete nucleotide sequence of the extensive PKD1 gene coding region. Further, PKD1 alternative splicing features are discussed, below.



   The coding region of the PKD1 gene is complex and extensive, containing a total of 46 exons and producing a transcript of approximately 14 kb in length. FIG. 5A depicts the structure of the PKD1 gene transcript. A total of 46 exons were identified within the PKD1 gene. Additionally,  sequence analysis from a number of cDNA clones reveals that the gene may have alternatively spliced forms. FIGS.   5B-5C    show a table of exons, listing exon sizes and indicating which cDNA clones contain nucleotide sequences corresponding to sequences present within specific exons.



   FIGS. 6A-6P depict the PKD1 nucleotide sequence.



  Specifically, the top line of FIGS. 6A-6P shows the nucleotide sequence of the entire PKD1 coding region (SEQ ID
NO: 1). The term   "PKD1    gene", as used herein, refers to (a) the nucleotide sequence depicted in FIGS. 6A-6P (SEQ ID NO: 1); (b) any DNA sequence that hybridizes to the complement of the nucleotide sequence depicted in FIGS. 6A-6P (SEQ ID NO: 1), under highly stringent conditions,   e.a.,    hybridization to filter-bound DNA in 0.5 M NaHPO4, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 650, and washing in   0.1xSSC/0.1k    SDS at 680C (Ausubel F.M. et al., eds., 1989, Current Protocols in
Molecular Biology, Vol.

  I, Green Publishing Associates, Inc., and John Wiley  & sons, Inc., New York, at p. 2.10.3) and which encodes a gene product functionally equivalent to the
PKD1 gene product (SEQ ID NO: 2) depicted in FIGS. 6A-6P and/or (c) any DNA sequence that hybridizes to the complement of the nucleotide sequence depicted in FIGS. 6A-6P (SEQ ID
NO: 1) under less stringent conditions, such as moderately stringent conditions,   e.a.,    washing in   0.2xSSC/0.1    SDS at 420C (Ausubel et al., 1989, supra), yet which still encodes a gene product functionally equivalent to the PKD1 gene product depicted in FIGS. 6A-6P (SEQ ID NO: 2).



   The term "functionally equivalent" as used herein can refer to: 1) a gene product or peptide having the biological function of the PKD1 gene product depicted in FIGS. 6A-6P and/or the biological function of a PKD1 peptide domain, as depicted in FIGS. 7A-7B and 8; 2) a gene product containing at least one PKD1 peptide domain as depicted in FIGS. 7A-7B and 8; or 3) a gene product having an 80% overall amino acid residue similarity to the PKD1 gene product depicted in FIGS.



  6A-6P. The term "functionally equivalent gene" as used herein can further refer a nucleotide sequence which encodes  a gene product of 1, 2 or 3, as described earlier in this paragraph.



   The invention also includes nucleic acid molecules, preferably DNA molecules, that hybridize to, and are therefore the complements of, the DNA sequences (a) through (c), in the preceding paragraph. Such hybridization conditions may be highly stringent or less highly stringent, as described above. In instances wherein the nucleic acid molecules are oligonucleotides ("oligos"), highly stringent conditions may refer, e.a., to washing in   6xSSC/0.05k    sodium pyrophosphate at 370C (for 14-base oligos),   48"C    (for 17-base   oligos) ,      55"C    (for 20-base   oligos) ,    and   60"C    (for 23-base oligos).

  These nucleic acid molecules may act as PKD1 antisense molecules, useful, for   exaL,ple,    in PKD1 gene regulation and/or as antisense primers in amplification reactions of PKD1 nucleic acid sequences. Further, such sequences may be used as part of ribozyme and/or triple helix sequences, also useful for PKD gene regulation. Still further, such molecules may be used as components of diagnostic methods whereby the level of PKD1 transcript may be deduced and/or the presence of an ADPKD-causing allele may be detected. Further, such sequences can be used to screen for and identify PKD1 homologs from, for example, other species.



   The invention also encompasses (a) DNA vectors that contain any of the foregoing coding sequences and/or their complements (i.e., antisense); (b) DNA expression vectors that contain any of the foregoing coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences; and (c) genetically engineered host cells that contain any of the foregoing coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences in the host cell.

  As used herein, regulatory   elements    include but are not limited to inducible and non-inducible promoters, enhancers, operators and other elements known to those skilled in the art that drive and regulate expression.  
For example, such regulatory elements may include CMV immediate early gene regulatory sequences, SV40 early or late promoter sequences on adenovirus, lac system,   trt    system, tac system or the trc system sequences. The invention includes fragments of any of the DNA sequences disclosed herein.



   In addition to the PKD1 gene sequences described above, homologs of the PKD1 gene of the invention, as may, for example be present in other, non-human species, may be identified and isolated by molecular biological techniques well known in the art and, for example, labelled probes of small as 12 bp. Further, mutant PKD1 alleles and additional normal alleles of the human PKD1 gene of the invention, may be identified using such techniques. Still further, there may exist genes at other genetic loci within the human genome that encode proteins which have extensive homology to one or more domains of the PKD1 gene product. Such genes may also be identified via such techniques.



   For example, such a previously unknown   PKD1-type    gene sequence may be isolated by performing a polymerase chain reaction (PCR; the experimental embodiment set forth by
Mullis, K.B., 1987, U.S. Patent No. 4,683,202) using two degenerate oligonucleotide primer pools designed on the basis of amino acid sequences within the PKD1 gene described herein (see,   e.a.    FIGS. 6A-6P, SEQ ID NO: 2). The template for the reaction may be cDNA obtained by reverse transcription of mRNA prepared from human or non-human cell lines or tissue known to express a PKD1 allele or PKD1 homologue. The PCR product may be subcloned and sequenced to insure that the amplified sequences represent the sequences of a PKD1 or a
PKD-like nucleic acid sequence. 

  The PCR fragment may then be used to isolate a full length PKD1 cDNA clone by radioactively labeling the amplified fragment and screening a bacteriophage cDNA library. Alternatively, the labeled fragment may be used to screen a genomic library. For a review of cloning strategies which may be used, see e.g.,
Maniatis, 1989, Molecular Cloning, A Laboratory Manual, Cold
Springs Harbor Press, N.Y.; and Ausubel et al., 1989, Current  
Protocols in Molecular Biology, (Green Publishing Associates and Wiley Interscience, N.Y.).



   5.2. THE PKD1 GENE PRODUCT
 The PKD1 gene products of the invention include the PKD1 gene product encoded by the PKD1 nucleotide sequence depicted in FIGS. 6A-6P (SEQ ID NO: 2). The PKD1 gene product shown in FIGS. 6A-6P is a protein of 4304 amino acid residues, with a predicted mass of approximately 467 kilodaltons. This PKD1 gene product contains as least five distinct peptide domains which are likely to be involved in protein-protein and/or protein-carbohydrate interactions. Further, this PKD1 gene product shares amino acid sequence similarity with a number of extracellular matrix proteins. (See FIGS. 7A-7B and 8, which list the PKD1 gene product domains.) The PKD1 gene product domains are more fully described below, in the
Example presented in Section 10.



   In addition, PKD1 gene products that represent functionally equivalent gene products are within the scope of the invention. "Functionally equivalent" as used herein is as defined in Section 5.1, above. Such an equivalent PKD1 gene product may contain deletions, additions or substitutions of amino acid residues within the PKD1 sequence encoded by the PKD1 gene sequences described, above, in
Section 5.1.3, but which result in a silent change thus producing a functionally equivalent PKD1 protein. Such amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipatic nature of the residues involved.

  For example, negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; amino acids with uncharged polar head groups having similar hydrophilicity values include the following: leucine, isoleucine, valine, glycine, analine, asparagine, glutamine, serine, threonine, phenylalanine and tyrosine. As used herein, a functionally equivalent PKD1 refers to a protein that exhibits substantially the same  biological activity as the   PKDl    gene product encoded by the
PKD1 gene sequences described in Section 5.1.1, above.



   PKDl gene products and peptides substantially similar to the PKD1 gene product encoded by the   PKDI    gene sequences described in Section 5.1, above, which cause ADPKD symptoms are also intended to fall within the scope of the invention.



  Such gene products and peptides may include dominant mutant
PKD1 gene products, or PKD1 gene products functionally equivalent to such mutant PKD1 gene products. By "functionally equivalent mutant   PKDl    gene product" it is meant PKDl-like proteins that exhibit a biological activity substantially similar to the activity demonstrated by dominant mutant PKDl gene products.



   The PKDl wild type or mutant protein may be purified from natural sources, as discussed in Section 5.2.1, below, or may, alternatively, be chemically synthesized or recombinantly expressed, as discussed in Section 5.2.2, below.



   5.2.1 PKD1 PROTEIN PURIFICATION METHODS
 The   PKDl    protein may be substantially purified from natural sources   (e.s.,    purified from cells) using protein separation techniques well known in the art. "Substantially purified" signifies purified away from at least about   90%    (on a weight basis), and from at least about 99% of other proteins, glycoproteins, and other macromolecules normally found in such natural sources.



   Such purification techniques may include, but are not limited to ammonium sulfate precipitation, molecular sieve chromatography, and/or ion exchange chromatography.



  Alternatively, or additionally, the PKD1 gene product may be purified by immunoaffinity chromatography using an immunoabsorbent column to which an antibody is immobilized which is capable of binding the PKD1 gene product. Such an antibody may be monoclonal or polyclonal in origin. If the
PKD1 gene product is specifically glycosylated, the glycosylation pattern may be utilized as part of a  purification scheme via, for example, lectin chromatography.



   The cellular sources from which the PKD1 gene product may be purified may include, but are not limited to, those cells that are expected, by Northern and/or Western blot analysis, to express the PKD1 gene.   Preferi;.y,    such cellular sources are renal tubular epithelial cells, bilary duct cells, skeletal muscle cells, whole brain cells, lung alveolar epithelial cell, and placental cell
 One or more forms of the PKD1 gene product may be secreted out of the cell, i.e., may be extracellular. Such extracellular forms of the PKD1 gene product may preferably be purified from whole tissue rather than cells, utilizing any of the techniques described above. Preferable tissue includes, but is not limited to those tissues than contain cell types such as those described above.

  Alternatively,
PKD1 expressing cells such as those described above may be grown in cell culture, under conditions well known to those of skill in the art. The PKD1 gene product may then be purified from the cell media using any of the techniques discussed above.



   5.2.2. PKD1 PROTEIN SYNTHESIS AND EXPRESSION METHODS
 Methods for the chemical synthesis of polypeptides   (e.a.,    gene products) or fragments thereof, are well-known to those of ordinary skill in the art,   e.a.,    peptides can be synthesized by solid phase techniques, cleaved from the resin and purified by preparative high performance liquid chromatography   (see,      e.s.,    Creighton, 1983, Proteins:
Structures and Molecular Principles, W.H. Freeman  & Co.,
N.Y., pp. 50-60). The composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing;   e.a.,    using the Edman degradation procedure (see   e.a.,    Creighton, 1983, supra at pp. 34-49). Thus, the PKD1 protein may be chemically synthesized in whole or in part.



   The PKD1 protein may additionally be produced by recombinant DNA technology using the PKD1 nucleotide sequences as described, above, in Section 5.1, coupled with  techniques well known in the art. Thus, methods for preparing the PKD1 polypeptides and peptides of the invention by expressing nucleic acid encoding PKD1 sequences are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing PKD1 protein coding sequences and appropriate transcriptional/translational control signals.



  These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. See, for example, the techniques described in Maniatis et al., 1989, Molecular
Cloning A Laboratory Manual, Cold Spring Harbor Laboratory,
N.Y. and Ausubel et al., 1989, Current Protocols in Molecular
Biology, Greene Publishing Associates and Wiley Interscience,
N.Y., both of which are incorporated by reference herein in their entirety. Alternatively, RNA capable of encoding PKD1 protein sequences may be chemically synthesized using, for example, automated or semi-automated synthesizers. See, for example, the techniques described in "Oligonucleotide
Synthesis", 1984, Gait, M.J. ed., IRL Press, Oxford, which is incorporated by reference herein in its entirety.



   A variety of host-expression vector systems may be utilized to express the PKD1 coding sequences of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, exhibit the PKD1 protein of the invention in situ. 

  These include but are not limited to microorganisms such as bacteria   (e.a.,    E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing PKD1 protein coding sequences; yeast   (e.s.,    Saccharomvces, Pichia) transformed with recombinant yeast expression vectors containing'the PKD1 protein coding sequences; insect cell systems infected with recombinant virus expression vectors   (e.a.,    baculovirus) containing the PKD1 protein coding  sequences; plant cell systems infected with recombinant virus expression vectors   te.a.,    cauliflower mosaic virus, CaMV;

   tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors   (e.a.,    Ti plasmid) containing the
PKD1 protein coding sequences coding sequence; or mammalian cell systems   (e.a.,    COS, CHO, BHK, 293, 3T3) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells   (e.a.,    metallothionein promoter) or from mammalian viruses   (e.a.,    the adenovirus late promoter; the vaccinia virus 7.5K promoter)
 In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the PKD1 protein being expressed.

  For example, when a large quantity of such a protein is to be produced, for the generation of antibodies or to screen peptide libraries, for example, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited to, the
E. coli expression vector pUR278 (Ruther et al., 1983, EMBO
J. 2:1791), in which the PKD1 protein coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye  & Inouye, 1985, Nucleic Acids Res. 13:31013109; Van Heeke  & Schuster, 1989, J. Biol. Chem. 264:55035509); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).

  In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned PKD1 protein can be released from the GST moiety.



   In an insect system,   AutoraDha    californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in   SDodoptera      fruqiperda     cells. The PKD1 coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).

  Successful insertion of PKD1 coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin   gene).    These recombinant viruses are then used to infect   SpodoPtera      fruginerda    cells in which the inserted gene is expressed   (e.a.,    see Smith et al., 1983, J.



  Viol. 46:584; Smith, U.S. Patent No. 4,215,051).



   In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the PKD1 coding sequence or interest may be ligated to an adenovirus transcription/translation control complex,   e.g.,    the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome   (e.a.,    region El or E3) will result in a recombinant virus that is viable and capable of expressing
PKD1 protein in infected hosts   (e.a.,    See Logan  & Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:3655-3659). Specific initiation signals may also be required for efficient translation of inserted PKD1 coding sequences.

  These signals include the ATG initiation codon and adjacent sequences. In cases where an entire PKD1 gene, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only a portion of the PKD1 coding sequence is inserted, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both  natural and synthetic.

  The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see
Bittner et al., 1987, Methods in Enzymol. 153:516-544).



   In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.a., glycosylation) and processing   (e.a.,    cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins.



  Appropriate cells lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, WI38, etc.



   For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the PKD1 protein may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements   (e.a.,    promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. 

  This method may advantageously be used to engineer cell lines which express the PKD1 protein. Such  engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the PKD1 protein.



   A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler, et al., 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska  & Szybalski, 1962, Proc.



  Natl. Acad. Sci. USA 48:2026), and adenine phosphoribosyltransferase (Lowy, et al., 1980, Cell 22:817) genes can be employed in   tk-,      hgprt    or apart cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler, et al., 1980, Natl. Acad. Sci. USA 77:3567; O'Hare, et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan  & Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin, et al., 1981, J. Mol. Biol. 150:1); and hygro, which confers resistance to hygromycin (Santerre, et al., 1984, Gene 30:147) genes.



   Whether produced by molecular cloning methods or by chemical synthetic methods, the amino acid sequence of the
PKD1 protein which may be used in the assays of the invention need not be identical to the amino acid sequence encoded by the PKD1 gene reported herein. The PKD1 proteins or peptides used may comprise altered sequences in which amino acid residues are deleted, added, or substituted, while still resulting in a gene product functionally equivalent to the
PKD1 gene product. "Functionally   equivalent,    as utilized herein, is as defined, above, in Section 5.1, and is additionally defined to refer to peptides capable of interacting with other cellular or extracellular molecules in a manner substantially similar to the way in which the corresponding portion of the endogenous PKD1 gene product would.



   For example, functionally equivalent amino acid residues may be substituted for residues within the sequence resulting  in a change of amino acid sequence. Such substitutes may be selected from other members of the class (i.e., non-polar, positively charged or negatively charged) to which the amino acid belongs;   e.a.,    the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and   glutamic;    the positively charged (basic) amino acids include arginine, lysine, and histidine; the negatively charged (acidic) amino acids include aspartic and glutamic acid.



   When used as a component in the assay systems described herein, the PKD1 gene product or peptide   (e.a.,    gene product fragment) may be labeled, either directly or indirectly, to facilitate detection of a complex formed between the PKD1 gene product and a test substance. Any of a variety of suitable labeling systems may be used including but not limited to radioisotopes such as   1251;    enzyme labelling systems that generate a detectable colorimetric signal or light when exposed to substrate; and fluorescent labels.



   Where recombinant DNA technology is used to produce the
PKD1 protein for the assay systems described herein, it may be advantageous to engineer fusion proteins that can facilitate labeling, immobilization and/or detection. For example, the coding sequence of the viral or host cell protein can be fused to that of a heterologous protein that has enzyme activity or serves as an enzyme substrate in order to facilitate labeling and detection. The fusion constructs should be designed so that the heterologous component of the fusion product does not interfere with binding of the host cell and viral protein.



   Indirect labeling involves the use of a third protein, such as a labeled antibody, which specifically binds to one of the binding partners,   i.e.,    either the PKD1 protein or its binding partner used in the assay. Such antibodies include but are not limited to polyclonal, monoclonal, chimeric,  single chain, Fab fragments and fragments produced by an Fab expression library.



   5.3. ANTIBODIES REACTIVE WITH PKD1 GENE PRODUCT
 Described herein are methods for the production of antibodies capable of specifically recognizing one or more
PKD1 gene product epitopes. Such antibodies may include, but are not limited to polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments,   F(ab')2    fragments, fragments produced by a FAb expression library, anti-idiotypic (anti
Id) antibodies, and epitope-binding fragments of any of the above. Such antibodies may be used, for example, in the detection of PKD1 gene product in a biological sample, or, alternatively, as a method for the inhibition of abnormal
PKD1 activity.

  Thus, such antibodies may be utilized as part of ADPKD treatment methods, and/or may be used as part of diagnostic techniques whereby patients may be tested for abnormal levels of PKD1 gene product, or for the presence of abnormal forms of the PKD1 protein.



   For the production of antibodies to PKD1, various host animals may be immunized by injection with PKD1 protein, or a portion thereof. Such host animals may include but are not limited to, rabbits, mice, and rats. Various adjuvants may be used to increase the immunological response, depending on the host species, including, but not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and   Corynebacteriumparoum.   



   Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of animals immunized with an antigen, such as PKD1, or an antigenic functional derivative thereof. For the production of polyclonal antibodies, host animals such as those described above, may  be immunized by injection with PKD1 protein supplemented with adjuvants as also described above.



   Monoclonal antibodies which are substantially homogeneous populations of antibodies to a particular antigen, may be obtained by any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler   and'Milstein    (1975, Nature 256:495-497; and U.S. Patent No. 4,376,110), the human B-cell hybridoma technique (Kosbor et al., 1983, Immunology Today 4:72; Cole et al., 1983, Proc. Natl. Acad. Sci. USA 80:20262030), and the EBV-hybridoma technique (Cole et al., 1985,
Monoclonal Antibodies And Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). Such antibodies may be of any immunoglobulin class, including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The hybridoma producing the mAb of this invention may be cultivated in vitro or in vivo.

  Production of high titers of mAbs in vivo makes this the presently preferred method of production.



   In addition, techniques developed for the production of "chimeric antibodies" (Morrison et al., 1984, Proc. Natl.



  Acad. Sci., 81:6851-6855; Neuberger et al., 1984, Nature, 312:604-608; Takeda et al., 1985, Nature, 314:452-454; U.S.

 

  Patent No. 4,816,567, which is incorported by reference herein in its entirety) by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a murine variable region and a human immunoglobulin constant region.



   Alternatively, techniques described for the production of single chain antibodies (U.S. Patent 4,946,778; Bird, 1988, Science 242:423-426; Huston et al., 1988, Proc. Natl.



  Acad. Sci. USA 85:5879-5883; and Ward et al., 1989, Nature   334:544-546)    can be adapted to produce PKD1-single chain antibodies. Single chain antibodies are formed by linking  the heavy and light chain fragment of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.



   Further, PKD1-humanized monoclonal antibodies may be produced using standard techniques (see, for example, U.S.



  Patent No. 5,225,539, which is incorporated herein by reference in its entirety)
 Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, such fragments include but are not limited to: the   F(ab')2    fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the   F(ab')2    fragments. Alternatively, Fab expression libraries may be constructed (Huse et al., 1989, Science,   246:1275-1281)    to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.



   5.4. SCREENING ASSAYS FOR COMPOUNDS
 THAT INTERACT WITH THE PKD1 GENE PRODUCT
 The following assays are designed to identify compounds that bind to the PKD1 gene product; other cellular proteins that interact with the PKD1 gene product; and compounds that interfere with the interaction of the PKD1 product with other cellular proteins.



   Compounds identified via assays such as those described herein may be useful, for example, in elaborating the biological function of the PKD1 gene product, and for ameliorating ADPKD symptoms caused by mutations within the
PKD1 gene. In instances whereby a mutation with the PKD1 gene causes a lower level of expression, and therefore results in an overall lower level of PKD1 activity in a cell or tissue, compounds that interact with the PKD1 gene product may include ones which accentuate or amplify the activity of the bound PKD1 protein. Thus, such compounds would bring about an effective increase in the level of PKD1 activity, thus ameliorating ADPKD symptoms.

  In instances whereby mutations with the PKD1 gene cause aberrant PKD1 proteins to  be made which have a deleterious effect that leads to ADPKD, compounds that bind PKD1 protein may be identified that inhibit the activity of the bound PKD1 protein.



   This decrease in the aberrant PKD1 activity can therefore, serve to ameliorate ADPKD symptoms. Assays for testing the effectiveness of compounds, identified by, for example, techniques such as those described in this Section are discussed, below, in Section 5.3.



   5.5. IN VITRO SCREENING ASSAYS FOR
 COMPOUNDS THAT BIND TO THE PKD1 PROTEIN
 In vitro systems may be designed to identify compounds capable of binding the PKD1 gene of the invention. Such compounds may include, but are not limited to, peptides made of D-and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam, K.S. et   al.,    1991,
Nature 354:82-84), phosphopeptides (in, for example, the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang, Z. et al., 1993, Cell 72:767-778), antibodies, and small or large organic or inorganic molecules.

  Compounds identified may be useful, for example, in modulating the activity of PKD1 proteins, preferably mutant PKD1 proteins, may be useful in elaborating the biological function of the PKD1 protein, may be utilized in screens for identifying compounds that disrupt normal PKD1 interactions, or may in themselves disrupt such interactions.



   The principle of the assays used to identify compounds that bind to the PKD1 protein involves preparing a reaction mixture of the PKD1 protein and the test compound under conditions and for a time sufficient to allow the two components to interact and bind, thus forming a complex which can be removed and/or detected in the reaction mixture.



  These assays can be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring
PKD1 or the test substance onto a solid phase and detecting   PKDl/test    substance complexes anchored on the solid phase at the end of the reaction. In homogeneous assays, the entire  reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested.



   In a heterogeneous assay system, the PKD1 protein may be anchored onto a solid surface, and the test substance, which is not anchored, is labeled, either directly or indirectly.



  In practice, microtiter plates are conveniently utilized.



  The anchored component may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the protein and drying. Alternatively, an immobilized antibody, preferably a monoclonal antibody, specific for the protein may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.



   In order to conduct the assay, the labeled component is added to the coated surface containing the anchored component. After the reaction is complete, unreacted components are removed   (e.a.,    by washing) under conditions such that any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the labeled compound is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the labeled component is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface;   e.a.,    using a labeled antibody specific for the binding partner (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).

 

   Alternatively, a heterogenous reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected;   e.a.,    using an immobilized antibody specific for PKD1 or the test substance to anchor any complexes formed in solution, and a labeled  antibody specific for the other binding partner to detect anchored complexes.



   In an alternate embodiment of the invention, a homogeneous assay can be used. In this approach, a preformed complex of the PKD1 protein and a known binding partner is prepared in which one of the components is labeled, but the signal generated by the label is quenched due to complex formation (see,   e.g.,    U.S. Patent No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays) . The addition of a test substance that competes with and displaces one of the binding partners from the preformed complex will result in the generation of a signal above background.



   5.6. ASSAYS FOR CELLULAR PROTEINS
 THAT INTERACT WITH PKD1 PROTEIN
 Any method suitable for detecting protein-protein interactions may be employed for identifying novel PKD1cellular or extracellular protein interactions. For example, some traditional methods which may be employed are co-immunoprecipitation, crosslinking and copurification through gradients or chromatographic columns. Additionally, methods which result in the simultaneous identification of the genes coding for the protein interacting with a target protein may be employed. These methods include, for example, probing expression libraries with labeled target protein, using this protein in a manner similar to antibody probing of   Agtll    libraries.



   One such method which detects protein interactions in vivo, the yeast two-hybrid system, is described in detail for illustration only and not by way of limitation. One version of this system has been described (Chien et al., 1991, Proc.



  Natl. Acad. Sci. USA,   88:9578-9582)    and is commercially available from Clontech (Palo Alto, CA).



   Briefly, utilizing such a system, plasmids are constructed that encode two hybrid proteins: one consists of the DNA-binding domain of a transcription activator protein fused to one test protein "X" and the other consists of the  activator protein's activation domain fused to another test protein "Y". Thus, either "X" or "Y" in this system may be wild type or mutant PKD1, while the other may be a test protein or peptide. The plasmids are transformed into a strain of the yeast Saccharomvces cerevisiae that contains a reporter gene   (e.a.,    lacZ) whose regulatory region contains the activator's binding sites. Either hybrid protein alone cannot activate transcription of the reporter gene, the DNAbinding domain hybrid because it does not provide activation function and the activation domain hybrid because it cannot localize to the activator's binding sites.

  Interaction of the two proteins reconstitutes the functional activator protein and results in expression of the reporter gene, which is detected by an assay for the reporter gene product.



   The two-hybrid system or related methodology can be used to screen activation domain libraries for proteins that interact with a PKDl protein. Total genomic or cDNA sequences are fused to the DNA encoding an activation domain.



  This library and a plasmid encoding a hybrid of the PKD1 protein fused to the DNA-binding domain are cotransformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene. These colonies are purified and the plasmids responsible for reporter gene expression are isolated. DNA sequencing is then used to identify the proteins encoded by the library plasmids.



   For example, and not by way of limitation, the PKD1 gene can be cloned into a vector such that it is translationally fused to the DNA encoding the DNA-binding domain of the GAL4 protein. A cDNA library of the cell line from which proteins that interact with PKD1 are to be detected can be made using methods routinely practiced in the art. According to this particular system, for example, the cDNA fragments can be inserted into a vector such that they are translationally fused to the activation domain of   GAL4.    This library can be co-transformed along with the PKD1-GAL4 DNA binding domain fusion plasmid into a yeast strain which contains a lacZ gene  driven by a promoter which contains GAL4 activation sequences.

  A cDNA encoded protein, fused to GAL4 activation domain, that interacts with PKD1 will reconstitute an active
GAL4 protein and thereby drive expression of the lacZ gene.



  Colonies which express lacZ can be detected by their blue color in the presence of X-gal. The cDNA can then be extracted from strains derived from these and used to produce and isolate the   PKDl-interacting    protein using techniques routinely practiced in the art.



   5.7. ASSAYS FOR COMPOUNDS THAT INTERFERE
 WITH PKD1/CELLULAR PROTEIN INTERACTION
 The PKD1 protein of the invention may, in vivo, interact with one or more cellular or extracellular proteins. Such cellular proteins are referred to herein as "binding   partners?1.    Compounds that disrupt such interactions may be useful in regulating the activity of the PKD1 protein, especially mutant PKD1 proteins. Such compounds may include, but are not limited to molecules such as antibodies, peptides, and the like described in Section 5.2.1. above.



   In instances whereby ADPKD symptoms are caused by a mutation within the PRD1 gene which produces PKD1 gene products having aberrant, gain-of-function activity, compounds identified that disrupt such interactions may, therefore inhibit the aberrant PKD1 activity. Preferably, compounds may be identified which disrupt the interaction of mutant PKD1 gene products with cellular or extracellular proteins, but do not substantially effect the interactions of the normal PKD1 protein. Such compounds may be identified by comparing the effectiveness of a compound to disrupt interactions in an assay containing normal PKD1 protein to that of an assay containing mutant PKD1 protein.



   The basic principle of the assay systems used to identify compounds that interfere with the interaction between the PKD1 protein, preferably mutant PKD1 protein, and its cellular or extracellular protein binding partner or partners involves preparing a reaction mixture containing the  
PKD1 protein and the binding partner under conditions and for a time sufficient to allow the two proteins to interact and bind, thus forming a complex. In order to test a compound for inhibitory activity, the reaction is conducted in the presence and absence of the test compound, i.e., the test compound may be initially included in the reaction mixture, or added at a time subsequent to the addition of PKD1 and its cellular or extracellular binding partner; controls are incubated without the test compound or with a placebo.

  The formation of any complexes between the PKD1 protein and the cellular or extracellular binding partner is then detected.



  The formation of a complex in the control reaction, but not in the reaction mixture containing the test compound indicates that the compound interferes with the interaction of the PKD1 protein and the interactive protein. As noted above, complex formation within reaction mixtures containing the test compound and normal PKD1 protein may also be compared to complex formation within reaction mixtures containing the test compound and mutant PKD1 protein. This comparison may be important in those cases wherein it is desirable to identify compounds that disrupt interactions of mutant but not normal PKD1 proteins.

 

   The assay for compounds that interfere with the interaction of the binding partners can be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring one of the binding partners onto a solid phase and detecting complexes anchored on the solid phase at the end of the reaction. In homogeneous assays, the entire reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested.

  For example, test compounds that interfere with the interaction between the binding partners,   e.s.,    by competition, can be identified by conducting the reaction in the presence of the test substance; i.e., by adding the test substance to the reaction mixture prior to or simultaneously with the PKD1 protein and interactive cellular or  extracellular protein. On the other hand, test compounds that disrupt preformed complexes,   e.a.    compounds with higher binding constants that displace one of the binding partners from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed.



  The various formats are described briefly below.



   In a heterogeneous assay system, one binding partner,   e.s.,    either the PKD1 protein or the interactive cellular or extracellular protein, is anchored onto a solid surface, and its binding partner, which is not anchored, is labeled, either directly or indirectly. In practice, microtiter plates are conveniently utilized. The anchored species may be immobilized by non-covalent or covalent attachments. Noncovalent attachment may be accomplished simply by coating the solid surface with a solution of the protein and drying.



  Alternatively, an immobilized antibody specific for the protein may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.



   In order to conduct the assay, the binding partner of the immobilized species is added to the coated surface with or without the test compound. After the reaction is complete, unreacted components are removed   (e.s.,    by washing) and any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the binding partner was pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the binding partner is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the binding partner (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).

  Depending upon the order of addition of reaction components, test compounds which inhibit complex formation or which disrupt preformed complexes can be detected.



   Alternatively, the reaction can be conducted in a liquid phase in the presence or absence of the test compound, the  reaction products separated from unreacted components, and complexes detected;   e.a.,    using an immobilized antibody specific for one binding partner to anchor any complexes formed in solution, and a labeled antibody specific for the other binding partner to detect anchored complexes. Again, depending upon the order of addition of reactants to the liquid phase, test compounds which inhibit complex or which disrupt preformed complexes can be identified.



   In an alternate embodiment of the invention, a homogeneous assay can be used. In this approach, a preformed complex of the PKD1 protein and the interactive cellular or extracellular protein is prepared in which one of the binding partners is labeled, but the signal generated by the label is quenched due to complex formation (see,   e.Ç.,    U.S. Patent
No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays) . The addition of a test substance that competes with and displaces one of the binding partners from the preformed complex will result in the generation of a signal above background. In this way, test substances which disrupt PKD1 protein-cellular or extracellular protein interaction can be identified.



   In a particular embodiment, the PKD1 protein can be prepared for immobilization using recombinant DNA techniques described in Section   5.1.2.2,    supra. For example, the PKD1 coding region can be fused to the glutathione-S-transferase (GST) gene using the fusion vector pGEX-SX-1, in such a manner that its binding activity is maintained in the resulting fusion protein. The interactive cellular or extracellular protein can be purified and used to raise a monoclonal antibody, using methods routinely practiced in the art and described above. This antibody can be labeled with the radioactive isotope   1251,    for example, by methods routinely practiced in the art. In a heterogeneous assay,   e.g.,    the GST-PKD1 fusion protein can be anchored to glutathione-agarose beads.

  The interactive cellular or extracellular protein can then be added in the presence or absence of the test compound in a manner that allows  interaction and binding to occur. At the end of the reaction period, unbound material can be washed away, and the labeled monoclonal antibody can be added to the system and allowed to bind to the complexed binding partners. The interaction between the PKD1 protein and the interactive cellular or extracellular protein can be detected by measuring the amount of radioactivity that remains associated with the glutathione-agarose beads. A successful inhibition of the interaction by the test compound will result in a decrease in measured radioactivity.



   Alternatively, the GST-PKD1 fusion protein and the interactive cellular or extracellular protein can be mixed together in liquid in the absence of the solid glutathioneagarose beads. The test compound can be added either during or after the binding partners are allowed to interact. This mixture can then be added to the glutathione-agarose beads and unbound material is washed away. Again the extent of inhibition of the binding partner interaction can be detected by adding the labeled antibody and measuring the radioactivity associated with the beads.



   In another embodiment of the invention, these same techniques can be employed using peptide fragments that correspond to the binding domains of the PKD1 protein and the interactive cellular or extracellular protein, respectively, in place of one or both of the full length proteins. Any number of methods routinely practiced in the art can be used to identify and isolate the protein's binding site. These methods include, but are not limited to, mutagenesis of one of the genes encoding the proteins and screening for disruption of. binding in a co-immunoprecipitation assay.

 

  Compensating mutations in the PKDl gene can be selected.



  Sequence analysis of the genes encoding the respective proteins will reveal the mutations that correspond to the region of the protein involved in interactive binding.



  Alternatively, one protein can be anchored to a solid surface using methods described in this Section above, and allowed to interact with and bind to its labeled binding partner, which  has been treated with a proteolytic enzyme, such as trypsin.



  After washing, a short, labeled peptide comprising the binding domain may remain associated with the solid material, which can be isolated and identified by amino acid sequencing. Also, once the gene coding for the for the cellular or extracellular protein is obtained, short gene segments can be engineered to express peptide fragments of the protein, which can then be tested for binding activity and purified or synthesized.



   For example, and not by way of limitation, PKD1 can be anchored to a solid material as described above in this sect ion by making a GST-PKD1 fusion protein and allowing it to bind to glutathione agarose beads. The interactive cellular protein can be labeled with a radioactive isotope, such as   35S,    and cleaved with a proteolytic enzyme such as trypsin. Cleavage products can then be added to the anchored
GST-PKD1 fusion protein and allowed to bind. After washing away unbound peptides, labeled bound material, representing the cellular or extracellular protein binding domain, can be eluted, purified, and analyzed for amino acid sequence by methods described in Section 5.1.2.2, supra. Peptides so identified can be produced synthetically or fused to appropriate facilitative proteins using recombinant DNA technology, as described in Section 5.1.2.2, supra.



   5.8. ASSAYS FOR ADKPD-INHIBITORY ACTIVITY
 Any of the binding compounds, including but not limited to, compounds such as those identified in the foregoing assay systems may be tested for anti-ADPKD activity. ADPKD, an autosomal dominant disorder, may involve underexpression of a wild-type PKD1 allele, or expression of a PKD1 gene product that exhibits little or no PKD1 activity. In such an instance, even though the PKD1 gene product is present, the overall level of normal PKD1 gene product present is insufficient and leads to ADPKD symptoms. As such, "anti
ADPKD activity", as used herein, may refer to a increase in the level of expression of the normal PKD1 gene product, to  levels wherein ADPKD symptoms are ameliorated. Additionally, the term may refer to an increase in the level of normal PKD1 activity in the cell, to levels wherein ADPKD symptoms are ameliorated.



   Alternatively, ADPKD may be caused by the production of an aberrant mutant form of the PKD1 protein, which either interferes with the normal allele product or introduces a novel function into the cell, which then leads to the mutant phenotype. For example, a mutant PKD1 protein may compete with the wild type protein for the binding of a substance required to relay a signal inside or outside of a cell.



  Circumstances such as these are referred to as "gain of function" mutations. It is possible that different mechanisms could be occurring in different patients which can lead to mutant phenotypic variations.



     tAnti-ADPKD    activity", as used herein, may refer to a decrease in the level and/or activity of such a mutant PKD1 protein so that symptoms of PKD1 are ameliorated.



   Cell-based and animal model-based assays for the identification of compounds exhibiting anti-ADPKD activity are described below.



   5.8.1. CELL BASED ASSAYS
 Cells that contain and express mutant PKD1 gene sequences which encode mutant PKD1 protein, and thus exhibit cellular phenotypes associated with ADPKD, may be utilized to identify compounds that possess anti-ADPKD activity. Such cells may include cell lines consisting of naturally occurring or engineered cells which express mutant or express both normal and mutant PKD1 gene products. Such cells include, but are not limited to renal epithelial cells, including primary and immortalized human renal tubular cells,
MDCK cells, LLPCKl cells, and human renal carcinoma cells.



   Cells, such as those described above, which exhibit
ADPKD-like cellular phenotypes, may be exposed to a compound suspected of exhibiting anti-ADPKD activity at a sufficient concentration and for a time sufficient to elicit such anti  
ADPKD1 activity in the exposed cells. After exposure, the cells are examined to determine whether one or more of the
ADPKD-like cellular phenotypes has been altered to resemble a more wild type, non-ADPKD phenotype.



   Among the cellular phenotypes which may be followed in the above assays are differences in the apical/basolateral distribution of membrane proteins. For example, normal (i.e., non-ADPKD) renal tubular cells in situ and in culture under defined conditions have a characteristic pattern of apical/basolateral distribution of cell surface markers.



  ADPKD renal cells, by contrast, exhibit a distribution pattern that reflects a partially reversed apical/basolateral polarity relative to the normal distribution. For example, sodium-potassium ATPase is found on the basolateral membranes of renal epithelial cells but is found on the apical surface of ADPKD epithelial cells, both in cystic epithelia in vivo and in ADPKD cells in culture (Wilson, et al., 1991, Am. J.



  Physiol. 260:F420-F430). Among the other markers which exhibit an alteration in polarity in normal versus ADPKD affected cells are the EGF receptor, which is normally located basolaterally, but in ADPKD cells is mislocated to the apical surface. Such a apical/basolateral marker distribution phenotype may be followed, for example, by standard immunohistology techniques using antibodies specific to the marker(s) of interest in conjunction with procedures that are well known to those of skill in the art.



   Additionally, assays for the function of the PKDl gene product can, for example, include a measure of extracellular matrix (ECM) components, such as proteoglycans, laminin, fibronectin and the like, in that studies in both ADPKD and in rat models of acquired cystic disease (Carone, F.A. et al., 1989, Kidney International 35:1034-1040) have shown alterations in such components. Thus, any compound which serves to create an extracellular matrix environment which more fully mimics the normal ECM should be considered as a candidate for testing for an ability to ameliorate ADPKD symptoms.  



   5.8.2 ANIMAL MODEL ASSAYS
 The ability of a compound, such as those identified in the foregoing binding assays, to prevent or inhibit disease may be assessed in animal models for ADPKD. Several naturally-occurring mutations for renal cystic disease have been found in animals. While these are not perfect models of
ADPKD, they provide test systems for assaying the effects of compounds that interact with PKD1 proteins. Of these models, the   Han:SPRD    rat model is the only autosomal dominant example. Such a model is well known to those of skill in the art. See, for example, Kaspareit-Rittinghausen et al., 1989,
Vet. Path. 26:195. In addition, several recessive models exist (Reeders, S., 1992, Nature Genetics 1:235).



   Additionally, animal models exhibiting ADPKD-like symptoms may be engineered by utilizing PKD1 sequences such as those described, above, in Section 5.1, in conjunction with techniques for producing transgenic animals that are well known to those of skill in the art.



   Animals of any species, including, but not limited to, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, goats, and non-human primates,   e.a.,    baboons, squirrels, monkeys, and chimpanzees may be used to generate such ADPKD animal models.

 

   In instances wherein the PKD1 mutation leading to ADPKD symptoms causes a drop in the level of PKD1 protein or causes an ineffective PKD1 protein to be made (i.e., the PKD1 mutation is a dominant loss-of-function mutation) various strategies may be utilized to generate animal models exhibiting ADPKD-like symptoms. For example, PKD1 knockout animals, such as mice, may be generated and used to screen for compounds which exhibit an ability to ameliorate ADPKD systems. Animals may be generated whose cells contain one inactivated copy of a PKD1-homologue. In such a strategy, human PKDl gene sequences may be used to identify a PKD1 homologue within the animal of interest, utilizing techniques described, above, in Section 5.1.

  Once such a PKD1 homologue has been identified, well-known techniques such as those  described, below, in Section 5.8.2.1. may be utilized to disrupt and inactivate the endogenous PKD1 homolog, and further, to produce animals which are heterozygous for such an inactivated PKD1 homolog. Such animals may then be observed for the development of ADPKD-like symptoms.



   In instances wherein a PKD1 mutation causes a PKD1 protein having an aberrant PKD1 activity which leads to ADPKD symptoms (i.e., the PKD1 mutation is a dominant gain-offunction mutation) strategies such as those now described may be utilized to generate ADPKD animal models. First, for example, a human PKD1 gene sequence containing such a gainof-function PKD1 mutation, and encoding such an aberrant PKD1 protein, may be introduced into the genome of the animal of interest by utilizing well known techniques such as those described, below, in Section 5.8.2.1. Such a PKD1 nucleic acid sequence must be controlled by a regulatory nucleic acid sequence which allows the mutant human PKD1 sequence to be expressed in the cells, preferably kidney cells, of the animal of interest. The human PKD1 regulatory promoter/enhancer sequences may be sufficient for such expression.

  Alternatively, the mutant PKD1 gene sequences may be controlled by regulatory sequences endogenous to the animal of interest, or by any other regulatory sequences which are effective in bringing about the expression of the mutant human PKD1 sequences in the animal cells of interest.



   Expression of the mutant human PKD1 gene may be assayed, for example, by standard Northern analysis, and the production of the mutant human PKD1 gene product may be assayed by, for example, detecting its presence by utilizing techniques whereby binding of an antibody directed against the mutant human PKD1 gene product is detected. Those animals found to express the mutant human PKD1 gene product may then be observed for the development of ADPKD-like symptoms.



   Alternatively, animal models of ADPKD may be produced by engineering animals containing mutations within one copy of their endogenous PKDl-homologue which correspond to gain-of  function mutations within the human PKD1 gene. Utilizing such a strategy, a PKD1 homologue may be identified and cloned from the animal of interest, using techniques such as those described, above, in Section 5.1. One or more gain-offunction mutations may be engineered into such a PKD1 homolog which correspond to gain-of-function mutations within the human PKD1 gene. By "corresponding", it is meant that the mutant gene product produced by such an engineered PKD1 homologue will exhibit an aberrant PKD1 activity which is substantially similar to that exhibited by the mutant human
PKD1 protein.



   The engineered PKD1 homologue may then be introduced into the genome of the animal of interest, using techniques such as those described, below, in Section 5.8.2.1. Because the mutation introduced into the engineered PKD1 homologue is expected to be a dominant gain-of-function mutation, integration into the genome need not be via homologous recombination, although such a route is preferred.



   Once transgenic animals have been generated, the expression of the mutant PKD1 homolog gene and protein may be assayed utilizing standard techniques, such as Northern and/or Western analyses. Animals expressing mutant PKD1 homolog proteins within the animals of interest, in cells or tissues, preferably kidney, of interest, the transgenic animals may be observed for the development of ADPKD-like symptoms.



   Any of the ADPKD animal models described herein may be used to test compounds for an ability to ameliorate ADPKD symptoms.



   In addition, as described in detail in Section 5.11 infra, such animal models can be used to determine the   LD50    and the   EDso    in animal subjects, and such data can be used to determine the in vivo efficacy of potential ADPKD treatments.  



   5.8.2.1 PRODUCTION OF PKD1 TRANSGENIC ANIMALS
 Any technique known in the art may be used to introduce a PKD1 gene into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to pronuclear microinjection (Hoppe, P.C. and Wagner,   T.E.,    1989, U.S. Pat. No. 4,873,191); retrovirus mediated gene transfer into germ lines (Van der Putten et al., 1985,
Proc. Natl. Acad. Sci., USA 82:6148-6152); gene targeting in embryonic stem cells (Thompson et al., 1989, Cell 56:313321); electroporation of embryos   (to,    1983, Mol Cell. Biol.



  3:1803-1814); and sperm-mediated gene transfer (Lavitrano et al., 1989, Cell 57:717-723); etc. For a review of such techniques, see Gordon, 1989, Transgenic Animals, Intl. Rev.



  Cytol. 115:171-229, which is incorporated by reference herein in its entirety)
 When it is desired that the PKD1 transgene be integrated into the chromosomal site of the endogenous PKD1, gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous PKD1 gene of interest are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of, the nucleotide sequence of the endogenous
PKD1 gene.



   Once the PKD1 founder animals are produced, they may be bred, inbred, outbred, or crossbred to produce colonies of the particular animal. Examples of such breeding strategies include but are not limited to: outbreeding of founder animals with more than one integration site in order to establish separate lines; inbreeding of separate lines in order to produce compound PKD1 transgenics that express the
PKD1 transgene at higher levels because of the effects of additive expression of each PKD1 transgene; crossing of heterozygous transgenic animals to produce animals homozygous for a given integration site in order to both augment expression and eliminate the possible need for screening of animals by DNA analysis; crossing of separate homozygous  lines to produce compound heterozygous or homozygous lines;

   breeding animals to different inbred genetic backgrounds so as to examine effects of modifying alleles on expression of the PKD1 transgene and the development of ADPKD-like symptoms. One such approach is to cross the PKD1 founder animals with a wild type strain to produce an   F1    generation that exhibits ADPKD symptoms, such as the development of polycystic kidneys. The   F1    generation may then be inbred in order to develop a homozygous line, if it is found that homozygous PKD1 transgenic animals are viable.



   The present invention provides for transgenic animals that carry the transgene in all their cells, as well as animals which carry the transgene in some, but not all their cells, i.e., mosaic animals. The transgene may be integrated as a single transgene or in concatamers,   e.a.,    head-to-head tandems or head-to-tail tandems.



   5.8.2.2. SELECTION AND CHARACTERIZATION
 OF THE PKD1 TRANSGENIC ANIMALS
 The PKD1 transgenic animals that are produced in accordance with the procedures detailed, above, in Section 5.8.2.1., should be screened and evaluated to select those animals which may be used as suitable animal models for   ADPKD.   

 

   Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to verify that integration of the transgene has taken place.



  The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal in situ hybridization analysis, and reverse transcriptase-PCR (rt-PCR). Samples of PKD1-expressing tissue, kidney tissue, for example, may be evaluated immunocytochemically using antibodies specific for the PKD1 transgene gene product.



   The PKD1 transgenic animals that express PKD1 mRNA or gene product (detected immunocytochemically, using antibodies  directed against PKD1 tag epitopes) at easily detectable levels should then be further evaluated histopathologically to identify those animals which display characteristic ADPKDlike symptoms. Such transgenic animals serve as suitable model systems for ADPKD.



   5.8.2.3. USES OF THE PKD1 ANIMAL MODELS
 The PKD1 animal models of the invention may be used as model systems for ADPKD disorder and/or to generate cell lines that can be used as cell culture models for this disorder.



   The PKD1 transgenic animal model systems for ADPKD may be used as a test substrate to identify drugs, pharmaceuticals, therapies and interventions which may be effective in treating such a disorder. Potential therapeutic agents may be tested by systemic or local administration.



  Suitable routes may include oral, rectal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections, to name a few. The response of the animals to the treatment may be monitored by assessing the reversal of disorders associated with ADPKD. With regard to intervention, any treatments which reverse any aspect of ADPKD-like symptoms should be considered as candidates for human ADPKD therapeutic intervention. However, treatments or regimens which reverse the constellation of pathologies associated with any of these disorders may be preferred. Dosages of test agents may be determined by deriving dose-response curves, as discussed in Section 5.11, below.



   In an alternate embodiment, the PKD1 transgenic animals of the invention may be used to derive a cell line which may be used as a test substrate in culture, to identify agents that ameliorate ADPKD-like symptoms. While primary cultures derived from the PKD1 transgenic animals of the invention may be utilized, the generation of continuous cell lines is  preferred. For examples of techniques which may be used to derive a continuous cell line from the transgenic animals, see Small et al., 1985, Mol. Cell Biol. 5:642-648.



   5.9. COMPOUNDS THAT INHIBIT EXPRESSION,
 SYNTHESIS OR ACTIVITY OF MUTANT
 PKDl ACTIVITY
 As discussed above, dominant mutations in the PKD1 gene that cause ADPKD may act as gain-of-function mutations which produce a form of the PKD1 protein which exhibits an aberrant activity that leads to the formation of ADPKD symptoms. A variety of techniques may be utilized to inhibit the expression, synthesis, or activity of such mutant PKD1 genes and gene products (i.e., proteins).



   For example, compounds such as those identified through assays described, above, in Section 5.4, which exhibit inhibitory activity, may be used in accordance with the invention to ameliorate ADPKD symptoms. Such molecules may include, but are not limited, to small and large organic molecules, peptides, and antibodies. Inhibitory antibody techniques are described, below, in Section 5.9.2.



   Further, antisense and ribozyme molecules which inhibit expression of the PKD1 gene, preferably the mutant PKD1 gene, may also be used to inhibit the aberrant PKD1 activity. Such techniques are described, below, in Section 5.9.1. Still further, as described, below, in Section 5.9.1, triple helix molecules may be utilized in inhibiting the aberrant PKD1 activity.



   5.9.1. INHIBITORY ANTISENSE, RIBOZYME
 AND TRIPLE HELIX APPROACHES
 Among the compounds which may exhibit anti-ADPKD activity are antisense, ribozyme, and triple helix molecules.



  Such molecules may be designed to reduce or inhibit mutant
PKD1 activity. Techniques for the production and use of such molecules are well known to those of skill in the art.  



   Antisense RNA and DNA molecules act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation. With respect to antisense
DNA, oligodeoxyribonucleotides derived from the translation initiation site,   e.a.,    between the -10 and +10 regions of the PKD1 nucleotide sequence of interest, are preferred.



   Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by a endonucleolytic cleavage. The composition of ribozyme molecules must include one or more sequences complementary to the target PKD1 mRNA, preferably the mutant PKD1 mRNA, and must include the well known catalytic sequence responsible for mRNA cleavage. For this sequence, see U.S. Pat. No.



  5,093,246, which is incorporated by reference herein in its entirety. As such, within the scope of the invention are engineered hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of RNA sequences encoding PKD1, preferably mutant
PKD1 proteins.



   Specific ribozyme cleavage sites within any potential
RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequence: GUA, GUU and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features, such as secondary structure, that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays.



   Nucleic acid molecules to be used in triplex helix formation should be single stranded and composed of deoxynucleotides. The base composition of these oligonucleotides must be designed to promote triple helix  formation via Hoogsteen base pairing rules, which generally require sizeable stretches of either purines or pyrimidines to be present on one strand of a duplex. Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC triplets across the three associated strands of the resulting triple helix. The pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand. In addition, nucleic acid molecules may be chosen that are purine-rich, for example, contain a stretch of guanidine residues.

  These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting In GGC triplets across the three strands in the triplex.



   Alternatively, the potential sequences that can be targeted for triple helix formation may be increased by creating a so called "switchback" nucleic acid molecule.



  Switchback molecules are synthesized in an alternating 5'-3',   3'-5    manner, such that they base pair with one strand of a duplex first and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.

 

   It is possible that the antisense, ribozyme, and/or triple helix molecules described herein may reduce or inhibit the translation of mRNA produced by both normal and mutant
PKD1 alleles. In order to ensure that substantial normal levels of PKD1 activity are maintained in the cell, nucleic acid molecules that encode and express PKD1 polypeptides exhibiting normal PKD1 activity may be introduced into cells which do not contain sequences susceptible to whatever antisense, ribozyme, or triple helix treatments. Such sequences may be introduced via gene therapy methods such as those described, below, in Section 5.5. Alternatively, it may be preferable to coadminister normal PKD1 protein into the cell or tissue in order to maintain the requisite level of cellular or tissue PKD1 activity.  



   Antisense RNA and DNA molecules, ribozyme molecules and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides and oligoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis.



  Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense
RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Alternatively, antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.



   Various well-known modifications to the DNA molecules may be introduced as a means of increasing intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences of ribo- or deoxy- nucleotides to the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the oligodeoxyribonucleotide backbone.



   5.9.2. ANTIBODIES THAT REACT WITH PKD1 GENE PRODUCT
 Antibodies that are both specific for mutant PKD1 gene product and interfere with its activity may be used. Such antibodies may be generated using standard techniques described in Section 5.3., supra, against the proteins themselves or against peptides corresponding to the binding domains of the proteins. Such antibodies include but are not limited to polyclonal, monoclonal, Fab fragments,   F(ab')2    fragments, single chain antibodies, chimeric antibodies, humanized antibodies etc.



   The PKD1 protein appears to be an extracellular protein.



  Therefore, any of the administration techniques described,  below in Section 5.11 which are appropriate for peptide administration may be utilized to effectively administer inhibitory PKD1 antibodies to their site of action.



   5.10 METHODS FOR RESTORING PKD1 ACTIVITY
 As discussed above, dominant mutations in the PKD1 gene that cause ADPKD may lower the level of expression of the
PKD1 gene or, alternatively, may cause inactive or substantially inactive PKD1 proteins to be formed. In either instance, the result is an overall lower level of normal PKD1 activity in the tissues or cells in which PKD1 is normally expressed. This lower level of PKD1 activity, then, leads to
ADPKD symptoms. Thus, such PKD1 mutations represent dominant loss-of-function mutations. Described in this Section are methods whereby the level of normal PKD1 activity may be increased to levels wherein ADPKD symptoms are ameliorated.



   For example, normal PKD1 protein, at a level sufficient to ameliorate ADPKD symptoms may be administered to a patient exhibiting such symptoms. Any of the techniques discussed, below, in Section 5.11, may be utilized for such administration. One of skill in the art will readily know how to determine the concentration of effective, non-toxic doses of the normal PKD1 protein, utilizing techniques such as those described, below, in Section 5.11.



   Additionally, DNA sequences encoding normal PKD1 protein may be directly administered to a patient exhibiting ADPKD symptoms, at a concentration sufficient to produce a level of
PKD1 protein such that ADPKD symptoms are ameliorated. Any of the techniques discussed, below, in Section 5.11, which achieve intracellular administration of compounds, such as, for example, liposome administration, may be utilized for the administration of such DNA molecules. The DNA molecules may be produced, for example, by recombinant techniques such as those described, above, in Section 5.1, and its subsections.



   Further, patients with these types of mutations may be treated by gene replacement therapy. A copy of the normal
PKD1 gene or a part of the gene that directs the production  of a normal PKD1 protein with the function of the PKD1 protein may be inserted into cells, renal cells, for example, using viral or non-viral vectors which include, but are not limited to vectors derived from, for example, retroviruses, vaccinia virus, adeno-associated virus, herpes viruses, bovine papilloma virus or additional, non-viral vectors, such as plasmids. In addition, techniques frequently employed by those skilled in the art for introducing DNA into mammalian cells may be utilized. For example, methods including but not limited to electroporation, DEAE-dextran mediated DNA transfer; DNA guns, liposomes, direct injection, and the like may be utilized to transfer recombinant vectors into host cells.

  Alternatively, the DNA may be transferred into cells through conjugation to proteins that are normally targeted to the inside of a cell. For example, the DNA may be conjugated to viral proteins that normally target viral particles into the targeted host cell. Additionally, techniques such as those described in Sections 5.1 and 5.2 and their subsections, above, may be utilized for the introduction of normal PKD1 gene sequences into human cells.



   The PKD1 gene is very large and, further, encodes a very large, approximately 14 kb, transcript. Additionally, the
PKD1 gene product is large, having 4304 amino acids, with a molecular weight of about 467 kD. It is possible, therefore, that the introduction of the entire PKD1 coding region may be cumbersome and potentially inefficient as a gene therapy approach. However, because the entire PKD1 gene product may not be necessary to avoid the appearance of ADPKD symptoms, the use of a "minigene" therapy approach (see,   e.a.,    Ragot,
T. et al., 1993, Nature 361:647; Dunckley, M.G. et al., 1993,
Hum. Mol. Genet. 2:717-723) can serve to ameliorate such
ADPKD symptoms.

 

   Such a minigene system comprises the use of a portion of the PKD1 coding region which encodes a partial, yet active or substantially active PKD1 gene product. As used herein, "substantially active" signifies that the gene product serves to ameliorate ADPKD symptoms. Thus, the minigene system  utilizes only that portion of the normal PKD1 gene which encodes a portion of the PKD1 gene product capable of ameliorating ADPKD symptoms, and may, therefore represent an effective and even more efficient ADPKD gene therapy than full-length gene therapy approaches. Such a minigene can be inserted into cells and utilized via the procedures described, above, for full-length gene replacement. The cells into which the PKD1 minigene are to be introduced are, preferably, those cells, such as renal cells, which are affected by ADPKD.

  Alternatively, any suitable cell can be transfected with a PKD1 minigene as long as the minigene is expressed in a sustained, stable fashion and produces a gene product that ameliorates ADPKD symptoms. Regulatory sequences by which such a PKD1 minigene can be successfully expressed will vary depending upon the cell into which the minigene is introduced. The skilled artisan will be aware of appropriate regulatory sequences for the given cell to be used. Techniques for such introduction and sustained expression are routine and are well known to those of skill in the art.



   A therapeutic minigene for the amelioration of ADPKD symptoms can comprise a nucleotide sequence which encodes at least one PKD1 gene product peptide domain, as shown in FIGS.



  7A-7B and 8. For example, such PKD1 peptide domains (the approximate amino acid residue positions of which are listed in parentheses after each domain name) can include a leucinerich repeat domain (72 to 94, or 97 to 119) and/or a cysteine-rich repeat domain (32 to 65), a C-type (calcium dependent) lectin protein domain (405 to 534), an LDL-A module (641 to 671), one or more PKD domains (282 to 353; 1032 to 1124; 1138 to 1209; 1221 to 1292; 1305 to 1377; 1390 to 1463; 1477 to 1545; 1559 to 1629; 1643 to 1715; 1729 to 1799; 1815 to 1884; 1898 to 1968; 1983 to 2058; 2071 to 2142), or at least one C-terminal domain (2160 to 4304)   (i.e.,    a peptide domain found in the C-terminal half of the
PKD1 gene product).

  Minigenes which encode such PKD1 gene products can be synthesized and/or engineered using the PKD1  gene sequence (SEQ ID NO:1) disclosed herein, and by utilizing the amino acid residue domain designations found in
FIGS. 7A-7B and 8.



   Among the ways whereby the PKDl minigene product activity can be assayed involves the use of PKD1 knockout animal models. Such animal models express an insufficient level of the PKD1 gene product. The production of such animal models may be as described above, in Section 5.8.2, and involves methods well known to those of skill in the art.



  PKD1 minigenes can be introduced into the PKD1 knockout animal models as, for example, described above, in this
Section. The activity of the minigene can then be assessed by assaying for the amelioration of ADKPD-like symptoms.



  Thus, the relative importance of each of the PKD peptide domains, individually and/or in combination, with respect to
PKD1 gene activity can be determined.



   Cells, preferably, autologous cells, containing normal
PKD1 expressing gene sequences may then be introduced or reintroduced into the patient at positions which allow for the amelioration of ADPKD symptoms. Such cell replacement techniques may be preferred, for example, when the PKD1 gene product is a secreted, extracellular gene product.



   5.11. PHARMACEUTICAL PREPARATIONS
 AND METHODS OF ADMINISTRATION
 The identified compounds that inhibit PKD1 expression, synthesis and/or activity can be administered to a patient at therapeutically effective doses to treat polycystic kidney disease. A therapeutically effective dose refers to that amount of the compound sufficient to result in amelioration of symptoms of polycystic kidney disease.



     5.11.1. EFFECTIVE    DOSE
 Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals,   e.a.,    for determining the
LD50 (the dose lethal to   50%    of the population) and the ED50   (the dose therapeutically effective in 50% of the population) . The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio   LD50/ED5.    Compounds which exhibit large therapeutic indices are Preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.



   The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the   EDso    with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the   Ices0    (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.

  Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. Additional factors which may be utilized to optimize dosage can include, for example, such factors as the severity of the ADPKD symptoms as well as the age, weight and possible additional disorders which the patient may also exhibit. Those skilled in the art will be able to determine the appropriate dose based on the above factors.



   5.11.2. FORMULATIONS AND USE
 Pharmaceutical compositions for use in accordance with the present invention may be formulated in conventional  manner using one or more physiologically acceptable carriers or excipients.



   Thus, the compounds and their physiologically acceptable salts and solvates may be formulated for administration by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.



   For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents   (e.s.,    pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers   (e.s.,    lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants   (e.s.,    magnesium stearate, talc or silica); disintegrants   (e.s.,    potato starch or sodium starch glycollate); or wetting agents   (e.s.,    sodium lauryl sulphate). The tablets may be coated by methods well known in the art.

  Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use.



  Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents   (e.a.,    sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents   (e.a.,    lecithin or acacia); non-aqueous vehicles   (e.a.,    almond oil, oily esters, ethyl alcohol or fractionated vegetable   oils);    and preservatives   (e.a.,    methyl or propyl-phydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.

 

   Preparations for oral administration may be suitably formulated to give controlled release of the active compound.



   For buccal administration the compositions may take the form of tablets or lozenges fdrmulated in conventional manner.  



   For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant,   e.a.,    dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of   e.c.,    gelatin, for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.



   The compounds may be formulated for parenteral administration by injection,   e.a.,    by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle,   e.a.,    sterile pyrogen-free water, before use.



   The compounds may also be formulated in rectal compositions such as suppositories or retention enemas,   e.s.,    containing conventional suppository bases such as cocoa butter or other glycerides.



   In addition to the formulations described previously, the compounds may also be formulated as a depot preparation.



  Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.  



   The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.



   5.12. DIAGNOSIS OF PKD1 ABNORMALITIES
 A variety of methods may be employed, utilizing reagents such as PKD1 nucleotide sequences described in Sections 5.1, and antibodies directed against PKD1 gene product or peptides, as described, above, in Section 5.1.3.



  Specifically, such reagents may be used for the detection of the presence of PKD1 mutations, i.e., molecules present in diseased tissue but absent from, or present in greatly reduced levels relative to, the corresponding non-diseased tissue.



   The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one specific PKD1 nucleic acid or   anti-PKD1    antibody reagent described herein, which may be conveniently used,   e.a.,    in clinical settings, to diagnose patients exhibiting
PKD1 abnormalities.



   Any tissue in which the PKD1 gene is expressed may be utilized in the diagnostics described below.



   5.12.1 DETECTION OF PKD-1 NUCLEIC ACIDS
 RNA from the tissue to be analyzed may be isolated using procedures which are well known to those in the art.



  Diagnostic procedures may also be performed in situ directly upon tissue sections (fixed and/or frozen) of patient tissue obtained from biopsies or resections, such that no RNA purification is necessary. Nucleic acid reagents such as those described in Section 5.1, and its subsections, may be used as probes and/or primers for such in situ procedures (Nuovo, G.J., 1992, PCR in situ hybridization: protocols and applications, Raven Press, NY).  



   PKD1 nucleotide sequences, either RNA or DNA, may, for example, be used in hybridization or amplification assays of biological samples to detect abnormalities of PKD1 expression;   e.a.,    Southern or Northern analysis, single stranded conformational polymorphism (SSCP) analysis including in situ hybridization assays, alternatively, polymerase chain reaction analyses. Such analyses may reveal both quantitative abnormalities in the expression pattern of the PKD1 gene, and, if the PKD1 mutation is, for example, an extensive deletion, or the result of a chromosomal rearrangement, may reveal more qualitative aspects of the
PKD1 abnormality.



   Preferred diagnostic methods for the detection of PKD1 specific nucleic acid molecules may involve for example, contacting and incubating nucleic acids, derived from the target tissue being analyzed, with one or more labeled nucleic acid reagents as are described in Section 5.1, under conditions favorable for the specific annealing of these reagents to their complementary sequences within the target molecule. Preferably, the lengths of these nucleic acid reagents are at least 15 to 30 nucleotides. After incubation, all non-annealed nucleic acids are removed. The presence of nucleic acids from the target tissue which have hybridized, if any such molecules exist, is then detected.

  Using such a detection scheme, the target tissue nucleic acid may be immobilized, for example, to a solid support such as a membrane, or a plastic surface such as that on a microtiter plate or polystyrene beads. In this case, after incubation, non-annealed, labeled nucleic acid reagents of the type described in Section 5.1 and its subsections are easily removed. Detection of the remaining, annealed, labeled nucleic acid reagents is accomplished using standard techniques well-known to those in the art.



   Alternative diagnostic methods for the detection of PKD1 specific nucleic acid molecules may involve their amplification,   e.a.,    by PCR (the experimental embodiment set forth in Mullis, K.B., 1987, U.S. Patent No. 4,683,202),  ligase chain reaction (Barany, F., 1991, Proc. Natl. Acad.



  Sci. USA 88:189-193), self sustained sequence replication (Guatelli, J.C. et al., 1990, Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh,   D.Y    et al., 1989, Proc. Natl. Acad. Sci. USA 86:1173-1177),
Q-Beta Replicase (Lizardi, P.M. et al., 1988, Bio/Technology 6:1197), or any other RNA amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of RNA molecules if such molecules are present in very low numbers.



   In one embodiment of such a detection scheme, a cDNA molecule is obtained from the target RNA molecule   (e.q.,    by reverse transcription of the RNA molecule into cDNA).



  Tissues from which such RNA may be isolated include any tissue in which wild type PKD1 is known to be expressed, including, but not limited, to kidney tissue and lymphocyte tissue. A target sequence within the cDNA is then used as the template for a nucleic acid amplification reaction, such as a PCR amplification reaction, or the like. The nucleic acid reagents used as synthesis initiation reagents   (e.a.,    primers) in the reverse transcription and nucleic acid amplification steps of this method are chosen from among the
PKD1 nucleic acid reagents described in Section 5.1 and its subsections. The preferred lengths of such nucleic acid reagents are at least 15-30 nucleotides. For detection of the amplified product, the nucleic acid amplification may be performed using radioactively or non-radioactively labeled nucleotides. 

  Alternatively, enough amplified product may be made such that the product may be visualized by standard ethidium bromide staining or by utilizing any other suitable nucleic acid staining method.



   5.12.2. DETECTION OF PKD1 GENE PRODUCT AND PEPTIDES
 Antibodies directed against wild type or mutant PKDl gene product or peptides, which are discussed, above, in  
Section 5.3, may also be used as ADPKD diagnostics, as described, for example, herein. Such diagnostic method, may be used to detect abnormalities in the level of PKD1 protein expression, or abnormalities in the location of the PKD1 tissue, cellular, or subcellular location of PKD1 protein.



  For example, in addition, differences in the size, electronegativity, or antigenicity of the mutant PKD1 protein relative to the normal PKD1 protein may also be detected.



   Protein from the tissue to be analyzed may easily be isolated using techniques which are well known to those of skill in the art. The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, "Antibodies: A
Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold
Spring Harbor, New York), which is incorporated herein by reference in its entirety.



   Preferred diagnostic methods for the detection of wild type or mutant PKDl gene product or peptide molecules may involve, for example, immunoassays wherein PKD1 peptides are detected by their interaction with an anti-PKD1 specific peptide antibody.



   For example, antibodies, or fragments of antibodies, such as those described, above, in Section 5.3, useful in the present invention may be used to quantitatively or qualitatively detect the presence of wild type or mutant PKD1 peptides. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody (see below) coupled with light microscopic, flow cytometric, or fluorimetric detection. Such techniques are especially preferred if PKD1 gene products or peptides are expressed on the cell surface.



   The antibodies (or fragments thereof) useful in the present invention may, additionally, be employed histologically, as in immunofluorescence or immunoelectron microscopy, for in situ detection of PKD1 gene product or peptides. In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto  a labeled antibody of the present invention. The histological sample may be taken from a tissue suspected of exhibiting ADPKD. The antibody   (or    fragment) is preferably applied by overlaying the labeled antibody (or fragment) onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the PKD1 peptides, but also their distribution in the examined tissue.



  Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order   t6    achieve such in situ detection.



   Immunoassays for wild type or mutant PKD1 gene product or peptides typically comprise incubating a biological sample, such as a biological fluid, a tissue extract, freshly harvested cells, or cells which have been incubated in tissue culture, in the presence of a detectably labeled antibody capable of identifying PKD1 peptides, and detecting the bound antibody by any of a number of techniques well-known in the art.



   The biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled PKD1 specific antibody.



  The solid phase support may then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on solid support may then be detected by conventional means.



   By "solid phase support or carrier" is intended any support capable of binding an antigen or an antibody. Wellknown supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention. The support material may have virtually  any possible structural configuration so long as the coupled molecule is capable of binding to an antigen or antibody.



  Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc.



  Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.



   The binding activity of a given lot of anti-wild type or mutant PKD1 peptide antibody may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.



   One of the ways in which the PKD1 peptide-specific antibody can be detectably labeled is by linking the same to an enzyme and use in an enzyme immunoassay   (EIA)    (Voller, A., "The Enzyme Linked Immunosorbent Assay (ELISA)", Diagnostic
Horizons 2:1-7, 1978) (Microbiological Associates Quarterly
Publication, Walkersville, MD); Voller, A. et al., J. Clin.



  Pathol. 31:507-520 (1978); Butler, J.E., Meth. Enzymol.



  73:482-523 (1981); Maggio, E. (ed.), ENZYME IMMUNOASSAY, CRC
Press, Boca Raton, FL, 1980; Ishikawa, E. et al., (eds.)
ENZYME IMMUNOASSAY, Kgaku Shoin, Tokyo, 1981). The enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alphaglycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and  acetylcholinesterase.

  The detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.



   Detection may be accomplished using any of a variety of other immunoassays. For example, by radioactively labeling the antibodies or antibody fragments it is possible to detect
PKD1 wild type or mutant peptides through the use of a radioimmunoassay (RIA)   (see,    for example, Weintraub, B.,
Principles of Radioimmunoassays, Seventh Training Course on
Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference   herein) .    The radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.



   It is also possible to label the antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin,   -phthaldehyde    and fluorescamine.

 

   The antibody can also be detectably labeled using fluorescence emitting metals such as   152Eu,    or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).



   The antibody also can be detect ably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol,  isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.



   Likewise, a bioluminescent compound may be used to label the antibody of the present invention.   Bioluminescence    is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.



   6. EXAMPLE: DETERMINATION OF THE PKD1 INTERVAL
 VIA GENETIC POLYMORPHISM ANALYSIS
 In the Working Example presented herein, genetic linkage studies are discussed which successfully reduced the potential PKD1 interval from approximately 750 kb to approximately 460 kb, thus substantially narrowing the genomic region in which the gene responsible for ADPKD lies.



   6.1 MATERIALS AND METHODS
Sequencina techniques: Sequencing of cDNA clones and genomic clones was carried out using an Applied Biosystems
ABI 373 automated sequencing machine according to the manufacturer's recommendations or by manual sequencing according to the method of Ausubel P. M. et al., eds., 1989,
Current Protocols in Molecular Biology, Vol. I, Green
Publishing Associates, Inc., and John Wiley  & Sons, New York, pp. 7.0.1  & ff.



   Inserts from the cDNA phage clones were excised with
EcoRI and ligated into the appropriate cloning sites in the polylinker of pBlueScript plasmid (Stratagene). Primers for sequencing of the plasmid clones were based on the known sequence of the polylinker. A second set of sequencing primers were based on the DNA sequences obtained from the first sequencing reactions. Sequences obtained using the second set of primers were used to design a third set of  primers and so on. Both strands of the double-stranded plasmids were sequenced.



   PCR products were sequenced using the dsDNA cycle sequencing system of GIBCO-BRL (Gaithersburg, MD) according to the manufacturer's instructions. PCR product was purified, prior to sequencing, by passing the DNA through a
Centricon column twice according to the manufacturer's instructions (Amicon, Beverly, MA, USA). 100-200ng of each purified PCR product was used as template in the sequencing reaction.



   Genomic sequences were obtained from PCR products as well as from subclones from the cosmids. To ensure the correct locus sequence was obtained over the duplicated locus. Only cGGG10 and cDEB11 sequence was utilized when identifiying intron/exon boundaries.



  DNA labelling: Double-stranded DNA probes were made by labelling DNA by the method of Feinberg and Vogelstein, 1983,
Anal. Biochem. 132: 6-13. Primers were end-labelled with   r32p-ATP    using the method of Ausubel F. M. et al., eds., 1989,
Current Protocols in Molecular Biology, Vol- 1, Green
Publishing Associates, Inc., and John Wiley  & Sons, New York, pp. 4.8.2  & f.



  PCR conditions: Conditions for the PCR reactions were determined empirically for each reaction by analyzing an array of reaction conditions with the following variables: magnesium concentrations of   lmM,    2mM, 4mM; annealing temperature; extension time; primer concentration and primer concentration ratio.



   The fixed conditions were:
 1. extension at 720C using Taq polymerase,   2.5u/100y1   
 reaction volume;
 2. denaturation at 950C for 1 minute; and
 3. annealing for 30 seconds.  



  Primer design: Primers were designed using the computer program "PRIMER".



  Genetic linkage studies: Genetic linkage studies were carried out using computerized algorithms (Lathrop GM., et al., 1984, Proc. Natl. Acad. Sci. USA,   81:3443-346;    Lathrop
GM and Lalouel J-M., 1984, Am. J. Hum. Genet. 36:460-465;
Lathrop G.M., Lalouel J.-M., Julier C., Ott J., 1985, Am.   ;   
Hum. Genet. 37:482-498).



  Single-stranded conformational   polymorhism    analysis (SSCP):
 SSCP analysis to detect sequence polymorphisms was carried out according to the method of Orita et al, 1989,
Genomics, 5:874-879. Primers were designed to amplify each exon (see figure 10 and Table 1, below). The 3' end of each primer was designed to lie   -20-50bp    from the nearest intron/exon boundary so that mutations in the splice donor and acceptor sites could be detected.



   Table 1: Primer Sequences from the PKD1 gene
EMI67.1     


<tb> Primer <SEP> Name <SEP> Sequence <SEP>    (5'-3')    <SEP> Sense/antisense
<tb>   KG8-F9    <SEP> CTGCCGGCCTGGTGTCG <SEP> sense
<tb> KG8-Fll <SEP> AGGGTCCACACGGGCTCGG <SEP> sense
<tb> KG8-F23 <SEP> CAGGGTGTCCGTGCGTGACTG <SEP> sense
<tb> KG8 <SEP> - <SEP> F2 <SEP> 5 <SEP> GTCCAGCACTCCTGGGGAGA <SEP> sense
<tb> KG8-F26 <SEP> ACGCAAGGACAAGGGAGTAG <SEP> sense
<tb>   KG8-F27    <SEP> AGTGCCGCGGCCTCCTGAC <SEP> sense
<tb>   KG8 <SEP> - <SEP> F2 <SEP> 8    <SEP> GCTGGCCTAGGCGGCTTCCA <SEP> sense
<tb>   KG8-MF2    <SEP> CACCCCACGGCTTTGCACT <SEP> i <SEP> sense
<tb> KG8-MF4 <SEP> CCCAGGCAG <SEP> CGAGGCTGTC <SEP> sense
<tb>   
EMI68.1     


<tb>   KG8 <SEP> -RO2    <SEP> ACACCAGGCCAACAGCGACTG <SEP> ant <SEP> 

   sense
<tb> KG8-R9 <SEP> ACAGCCACCAGGAGCAGGCTG <SEP> antisense
<tb>  <SEP> A
<tb> KG8-R13 <SEP> TGTAGCGCGTGAGCTCCAG <SEP> antisense
<tb> KG8-R23 <SEP> CACCCCACCCTACCCCAG <SEP> antisense
<tb> KG8-R24 <SEP> GGAGGCCACAGGTGAGGCT <SEP> ant <SEP> sense
<tb>   KGB <SEP> -R27    <SEP> CGGAGGAGTGAGGTGGGCTCC <SEP> antisense
<tb> KG8-R28 <SEP> AGCCATTGTGAGGACTCTCCC <SEP> antisense
<tb> NKG9-F2 <SEP> AAGACCTGATCCAGCAGGTCC <SEP> sense
<tb> NKG9-F07 <SEP> CAGCACGTCATCGTCAGG <SEP> sense
<tb> NKG9-R03 <SEP> CTCCCAGCCACCTTGCTC <SEP> antisense
<tb> NKG9-R07 <SEP> GCAGCTGTCGATGTCCAG <SEP> antisense
<tb> NKG9-RM2 <SEP> TCTGTCCAACAAAGGCCTG <SEP> antisense
<tb> 
 6.2 RESULTS
 It was previously shown that the PKD1 gene maps, by genetic linkage,

   to the interval between the polymorphic genetic markers D16S259 (which lies on the telomeric side of
PKD1) and D16S25 (which lies on the centromeric side of   PKD1)    (see Somlo et al., 1992, Genomics 13:152). The smallest interval between genetic markers, called the PKD1 interval was found to be approximately 750kb (see Germino et al., 1992, Genomics 13:144). The PKD1 interval was isolated as a series of forty overlapping cosmid and phage clones. The cloned DNA contained the entire PKD1 interval with the exception of two gaps of less than   iOkb    and less than 50kb (see FIG. 1; Germino et al., Genomics 13:144, 1992).

 

   In the Example presented herein, in order to reduce the
PKD1 interval still further, a systematic search for additional polymorphic markers was undertaken. Singlestranded DNA probes   (CA)8-15    were hybridized to the set of  clones from the PKD1 interval. The phage clone w5.2 (see
FIG. 1) was found to hybridize to the probe and the sequence flanking the (CA)n   (w5.2    repeat) was determined using phage
DNA as a template. Primers for the polymerase chain reaction (PCR) were designed and used to detect polymorphism within the w5.2Ca repeat. The position of the w5.2Ca repeat is shown in FIG. 2. This w5.2Ca repeat was used in genetic linkage studies in 15 PKD1 families and found to lie proximal to the PKD1 locus. This experiment reduced the size of the
PKD1 interval to approximately 460kb, as shown in FIG. 2.



   7. EXAMPLE: IDENTIFICATION OF POTENTIAL PKD1
 TRANSCRIPTS
 In the Working Example presented herein, transcription units within the 460 kb PKD1 interval, (FIG.2) defined in
Section 6, above, were identified. The interval was found to have a maximum of 27 transcriptional units (TU), which contained a total of approximately 300 kb.



   7.1 Materials and Methods cDNA librarv screening: cDNA libraries were prepared from several sources including EBV transformed lymphocytes, teratocarcinoma tissue, fetal kidney and HeLa cells. In addition a human adult kidney library was purchased from
Clontech Inc. (San Diego, CA).



   Total RNA from each tissue was prepared by the guanidinium chloride method. First strand cDNA synthesis was prepared using random six base oligonucleotides by the method of Zhou et al, Journal Biol. Chem., 267:12475 (1992). EcoRI sites within the cDNA were blocked by DNA methylase. The cDNA was flush-ended with T4 kinase and EcoRI linkers were added with DNA ligase. The cDNA was cleaved with EcoRI and ligated into either bacteriophage lambda-gtlO or lambda-ZAP (Stratagene). The phage were packaged with high-efficiency packaging extract (Stratagene). At least one million primary clones were plated. The library was amplified 100-fold and stored at   4"    C.  



   At least 500,000 plaques of each library were screened with each cosmid clone at a density of 25,000 per 75mm diameter plate. Duplicate filter lifts were made of each plate (Ausubel, supra). The radiolabelled probes were incubated with an excess of unlabelled denatured human DNA and then added to the library filters in a sodium phosphate buffer at 650 C. for 16 hours. The filters were washed in 2xSSC at   65"    C. for 1 hour and 0.1xSSC, 0.1xSDS at 650 C. for one hour. Kodak XAR-5 was exposed to the library filters for 4-16 hours. Duplicate positives were picked and replated at a density of approximately 100-500 per plate. Filter lifts of these secondary plates were made and hybridized as for the primary lifts; pure isolated plaques were obtained and inoculated into   50ml    cultures and the phage DNA was purified.



     Sequenceing    techniques: Techniques were as described in
Section 6.1, above.



   7.2 Results
 To identify transcribed sequences within the PKD1 interval (FIG. 2), the cosmid and phage clones from the interval were hybridized to cDNA libraries made from a variety of human tissues including fetal and adult kidney, teratocarcinoma, adult liver, lymphoblast, HeLa, and adult brain. More than 100 hybridizing cDNA clones were identified. These clones were subcloned into pBlueScript plasmids and sequenced. The sequence data combined with hybridization data (between cDNA clone and genomic clone) allowed the cDNA clones to be assigned to a maximum of 27 transcription units, as described below.



   Namely, hybridization between two cDNA clones was evidence that the clones are part of the same transcription units. Similarly, sequence identities of greater than 25bp between the cDNA clones were used as evidence that the clones were part of the same transcription unit.



   Table 2, below, lists these units (a-z, aa) by the name of the longest clone.  



   Table 2
 Putative Transcriptional Unit
 Sequences Isolated From the PKD1 Region
 CANDIDATE GENES IN THE   PKD1    REGION
 Insert
 Size
 Clone (kb) cDNA Libraries Motif
 a. 20.7 2.1 cy, terat
 b. SazD 2.7 cy G-protein ss subunit-like
 c. SazB 2.2 cy, terat scERV from yeast
 d. Saz10 4.0   cy    lym
 e. Sazl3 1.5 cy, terat tandem 120 amino-acid
 repeat; ZOl - family
 f. Saz20 5.5 cy,   lym.    terat
 g. KG8 3.4 lym
 h. NKG9 1.8 lym
 i. NKGIO 2.8 lym
 j. NKG11 2.4 lym
 k. Nik4 0.9 kid
 1. Nik7 2.3   lym,    terat rab gene motif
 m. KG3 3.8 terat, cy   G-protein    ss subunit-like
 n. Nik9 2.2 cy ankyrin repeat
 o. KG4 0.6 kid
 p. KM17 1.6 terat, cy   G-protein    ss subunit-like
 q. NiklO 1.6 lym
 r. KG5 2.6 cy zinc-finger protein
 s. KGI 1.1 kid DNase
 t.

  KG6 3.4 kid, cy, lym human homolog of
 mouse RNSP1 gene
 u. Nik3 3.2 terat, lym, cy    *   
 v. Nik2 3.4 terat,   lym,    cy *
 w. Nikl 0.8 kid *
 x. Nik8 1.6 lym *
 y. KG17 2.2 lym
 z.   All    1.4 cy cyclin-F homolog
 aa. MAR1 2.0 kid MDR-like * u, v, w, x are part of an 8kb transcriptional unit (nik 823) which produces a MDR-like channel.



  MAR1 is another member of the gene family.   ATPdependent    transporter cyclin protonchannel of vacuolar proton ATPase cDNA library from which the clone was obtained: cy=cyst; terat=teratocarcinoma;   lym = lymphoblast;    kid = kidney  
Thus, these 27 transcription units were considered by virtue of their genomic localization to be candidate genes for PKD1.



  The total transcribed cDNA in the 27 transcription units equalled about 60kb.



   The sequence of each clone was compared with sequences deposited in the public databases Genbank, EMBL, and
SwissProt. Several of the cDNA clones contained sequences predicted to code for known protein motifs. Because so little was known of the molecular basis of ADPKD none of the candidate genes could be ruled out by virtue of sequence motifs.



   8. PKD1 INTERVAL NORTHERN ANALYSIS
 In the Working Example presented herein, an analysis of the transcriptional expression patterns of the TUs described, above, in Section 7, was conducted.



   8.1 MATERIALS AND METHODS
Northern blot analysis: Poly A+ RNA   (2yg)    from heart, brain, placenta, lung, liver, skeletal muscle, kidney and pancreas was hybridized with radio-labelled cDNA probes from the TUs within the PKD1 interval, under standard conditions.

 

   8.2 RESULTS
 Inserts from the cDNA clones of the TUs described in
Section 7,and listed in Table 2, above, were used to probe
Northern blots containing total RNA and polyA-enriched RNA from normal human organs and from between 8 and 10 kidneys removed from patients with ADPKD.



   The expression profile was compared with the pattern of pathology in ADPKD to determine a priority for further characterization. The Northern analysis demonstrated that 26 of the TUs in the PKDl interval were expressed in kidney, with the exception of Nik9. Nik9 mRNA was found to be abundant in human brain but expressed at very low level in fetal and adult human kidney. These data, therefore, indicated that Nik9 is not the PKD1 gene. No consistent  differences were observed between normal and ADPKD kidneys for any transcript.



   9. EXAMPLE: PKD1 INTERVAL MUTATION SCREENS
 A systematic search was undertaken to detect mutations in ADPKD patients in the transcribed regions listed in Table 2. The mutation screen used several independent techniques.



  Southern blot analysis of patient DNA digested with at least three different restriction endonucleases was performed.



  Several differences between the restriction patterns were detected but none was found only in patients with ADPKD.



  Single-stranded conformational polymorphism analysis was carried out using cDNA isolated from patient transformed lymphocytes as a template. A large number of allelic differences was found but none were found to alter the deduced product of transcription. Sequence analysis of the
KG5 cDNA was carried out in seven ADPKD patients and one normal. The deduced coding region of 2.6kb was sequenced using cDNA, made by reverse transcription from patient transformed lymphocyte mRNA, as a template. The cDNA was amplified by PCR in a series of overlapping sections and the
PCR products were sequenced. No sequence differences were detected between patients and normal individuals. In this way more than   80k    of the coding DNA in the transcription units was scanned and no mutations were found in PKD1 patients.

  These experiments excluded the scanned segments of the transcription units with a likelihood of   95%    based on the reasonable assumption that no ADPKD mutation accounts for    > 70    of all ADPKD cases.



   Thus, the following transcription units were excluded: sazB, sazD sazl3, KG3, KG5, KGI,   saz20,    KM17, Nikl, Nik2,
Nik3,   Nicks,      KG17,    Nik7,   MAR1.    These excluded transcripts represent    > 80    of the combined identified coding sequences in the PKD1 region.



   It has previously been noted that de novo mutation to
ADPKD accounts for at least 1% of cases. Two mechanisms have been shown to account for the vast majority of new mutation  rates of this order. First, the coding region may be large.



  Duchenne muscular dystrophy (DMD) provides an example of this situation: the dystrophin gene which is mutated in DMD has a transcript of approximately   14kb.    About 30% of DMD cases arise by de novo mutation. The second mechanism that may account for a high new mutation rate is the presence of an unstable repetitive element. Unstable trinucleotide repeats in which the repeat sequence contains    > 50k      C   and G have been shown to cause the fragile X syndrome, Huntington's disease and myotonic dystrophy. In two of these diseases, high mutation rates or the appearance of progressively more severe disease in successive generations (anticipation) have been documented.



   A systematic search for trinucleotide repeats in the
PKD1 interval was undertaken. Single-stranded probes (15-25 nucleotides) containing all possible combinations of trinucleotide repeats were synthesized, radiolabelled and hybridized to Southern blots containing the complete set of clones comprising the PKD1 interval. The hybridization and washing conditions were adjusted to allow detection of all perfect repeats of 15 nucleotides or more. Eight separate banks of trinucleotide repeats within the PKD1 interval were found. Primers were designed so that the trinucleotide repeat arrays could be amplified by PCR and size-fractionated on polyacrylamide gels. No differences were found between
ADPKD patients and controls.



   Additionally, two other screening methods were attempted for the identification of trinucleotide expansions in the
PKD1 interval. Southern blots of DNA from normal and affected individuals was probed with inserts containing the repeats. This revealed no polymporphisms. Further, multiply restricted DNA samples (Rsa/Sau3A/Hinfl) samples were probed with trinucleotide repeat oligonucleotides. Though myotonic dystrophy and fragile-X mental retardation patients could be identified via such methods, it was not possible to identify any common pattern in ADPKD patients.  



   The cDNA clones Nikl, Nik2, Nik3, and Nik8 were found to hybridize to an 8kb transcript present in kidney. These clones were assumed to be part of the same transcript. PCR product that bridged the three gaps in sequence between the four clones were obtained using primers based on sequences within the four cDNA clones. In this way approximately 8kb of the transcribed DNA sequence of the gene represented by Nikl,
Nik2, Nik3, and Nik8 was obtained. Because the coding region is large the gene was expected to have a high spontaneous mutation rate and therefore to be a good candidate for the
PKD1 gene. A detailed exon-by-exon search of the gene, however, revealed no evidence of mutations in ADPKD patients.



  This left only one TU within the region which was considered large enough to be a reasonable candidate for the PKD1 gene.



  The characterization of clones and sequences within this TU, part of the putative PKDl gene, is described, below, in the
Working Examples presented in Sections 10 and 11.



   10. EXAMPLE: SSCP Analvsis of ADPKD Patients
 In the Working Example presented herein, an SSCP analysis of genomic DNA amplified from DNA derived from normal and ADPKD patients was conducted which identified
ADPKD-specific allelic differences which map to the single gene of the PKD1 interval which was described, above, in the
Working Example presented in Section 10.

 

   10.1 Materials and Methods
SSCP Analvsis: Single-Stranded Conformational Analysis (SSCP) was performed as follows: 50ng of genomic DNA was amplified by PCR under standard conditions in a reaction volume of 20   Zl.    Ten microliters of the amplified product was added to 90   Hl    of formamide buffer, heated at 970C for 4-5 minutes, and cooled on ice. Four microliters of the reaction mixture was loaded on a polyacrylamide gel (10%, 50:1 acrylamide:bisacrylamide) containing 10% glycerol. The gel was run at 40C for 12 hours with 10W power in 0.5 X TBE  buffer. The gel was dried and exposed to a Molecular Dynamic
Phosphor-Imager screen for 4 to 16 hours.



  Intron/Exon   Mating:    Primers produced from cDNA clones were used to PCR amplify genomic DNA sequences. Amplified products were sequenced, using standard methods. Those sequences which differed from the cDNA sequences indicated intron sequences.



  PCR Amplification: Procedures for amplification were as described, above, in Section 6.1.



   10.2 Results
 Because the large size of the putative
KG8/NKG9/NKG10/NKG11 transcript makes it a likely site for mutation, the intron/exon structure of part of the gene represented by KG8 and NKG9 was determined so that an exonby-exon search for mutations could be conducted. The exon/intron structure analysis allowed PCR primers to be designed for the amplification of several exons of the PKD1 gene.



   These primers were used to PCR-amplify genomic DNA and to perform SSCP of ADPKD patients and normal individuals. In two ADPKD patients SSCP patterns were observed that showed allelic differences. Both patients were heterozygous for an
SSCP variant that was not seen in a large number of normals from the normal population (Fig 3A-3B). In samples from these two individuals, 4 bands are visible, instead of the 2 single-strand bands seen in samples from normal individuals.



  The 4 bands are of equal intensity and are presumed to comprise two allelic sense strand and two allelic antisense strands.



   Thus, the results discussed in this Example, coupled with the analyses reported, above, in the Examples presented in Sections 6 through 9 provide positive correlative evidence that the gene corresponding to the putative transcription  unit of which the clones KG8, NKG9,   NKG10    and NKG11 are believed to be a part, is the PKD1 gene.



   11. EXAMPLE: MOLECULAR CHARACTERIZATION OF THE PKD1 GENE
 In this Example, the complex structure of the PKD1 gene and gene product is described. Included herein is a description of the PKD1 gene structure, the nucleotide sequence of the entire coding region of the PKD1 transcript, as well as the amino acid sequence and domain structure of the PKD1 gene product. This description not only represents the first elucidation of the entire PKD1 coding sequence, but additionally also corrects errors in the portion of the PKD1 coding region which had previously been reported. Also, a
ADPKD-1 causing mutation within the PKD1 gene which results in a frameshift is identified. Further, the strategy utilized to characterize this extensive and difficult nucleic acid region is summarized.



   A portion of the nucleotide sequence corresponding, in large part, to the 3' end of the PKD1 gene had recently been reported (European Polysystic Kidney Disease Consortium [hereinafter abbreviated EPKDC], 1994, Cell 77:881-894).



  Specifically, the terminal 5.6 kb of the PKD1 transcript were studied and an open reading frame of 4.8 kb was reported.



  The peptide this putative open reading frame encodes, which would correspond to the carboxy terminal portion of the PKD1 protein, did not reveal any homologies to known proteins and, if this derived amino acid sequence was, in fact, part of the
PKD1 protein, its sequence did not suggest a function for the
PKD1 gene product.



   For this lack of revealing information, in addition to the fact that only a small percentage of ADPKD-causing mutations appear to reside within the 3' end of the PKD1 gene, the characterization of the 5' end of the gene and a more complete analysis of the PKD1 gene and gene product were greatly needed.



   As acknowledged by the EPKDC (EPKDC, 1994, Cell 77:881894), however, the elucidation of the complete PKD1 coding  sequence presents major problems. Unlike the 3' end of the
PKD1 gene, the 5' two-thirds of the gene appear to be duplicated several times at other genomic positions.



  Further, at least some of these duplications are transcribed.



  Thus, great difficulties arise when attempting to distinguish sequence derived from the authentic PKD1 locus apart from sequence obtained from the duplicated PKD1-like loci.



   11.1. MATERIALS AND METHODS
 11.1.1. GENOMIC CLONES
 The human P1 phage named PKD 1521 was isolated from a human P1 library using primers from the adjacent TSC2 gene.



  The first screen utilized primers F33tcttctccaacttcacggctg,
R32aaccagccaggttttggtcct, followed by F38caagtccagctcctctccc,
R40gctctttaaggcgtccctc and ultimately screened with primers in the KG8 gene (F9/R5) see page 68 for KG8-R5 5' primer, while KG8-R5 5' gcgctttgcagacggtaggog 3'. The cosmid cGGG10 has been previously described (Germino, G.G., Weinstat
Saslow, D., Himmelbauer, H., Gillespie G.A.J., Somlo, S.,
Wirth, B., Barton, N., Harris, K.L., Frischauf,   A.M.    and
Reeders,   S.T.    (1992) Genomics, 13:144-151). The cosmid cGGG10 was mapped using various restriction enzymes as described by the manufacturers. A random library of the cosmid was constructed by cloning sheared DNA fragments into the SmaI site of pUC 19.

  Initial sequence assembly for the cosmid cGGG10 was performed on forward and reverse sequences of approximately 1000 random cloned fragments and a preliminary map was constructed using the restriction map of the cosmid. Directed subclones of cGGG10 were made in the plasmid pBluescript (Stratagene) in order to create sequencing islands specific physical locations. These large subclones from cGGG10 were then restricted with more frequent cutter enzymes and cloned into M13mpl9 and mpl8. In addition, if gaps were found in cloned regions, directed sequencing was performed from the flanking regions, to join the anchored contigs.

  A contig of 34.3 Kb was constructed, with two gaps in what appear to be highly repetitive regions  with no identifiable coding sequence. cDEB11 was has been described previously (Germino, G.G., Weinstat-Saslow,   D.,   
Himmelbauer, H., Gillespie G.A.J., Somlo, S., Wirth, B.,
Barton, N., Harris, K.L., Frischauf,   A.M.    and Reeders, S.T.



  (1992) Genomics, 13:144-151). A random library was constructed with sheared cDEB11 DNA and cloned into the SmaI site of pUC19. This cosmid was sequenced to obtain at least 2-fold coverage.



   The sequencing was done by cycle sequencing and run on
ABI machines following the manufacturer's instructions with modifications as described below. Because of the difficulty of sequencing certain regions, the standard chemistry of sequencing used withthe ABI machines had to be modified.



  Both dye terminator and dye primer sequence were used when appropriate with sequencing different regions. Different polymerases and different melting and polymerization conditions were also used in order to optimize the quality of the sequence. When sequencing across the CpG island at the 5' end of the PKD1 gene, the best sequencing results were obtained when adding 5% DMSO to the polymerization step and sequencing single-stranded templates.

 

   11.1.2. cDNA LIBRARY SCREENING
 The first cDNA used to screen libraries was KG8, which maps to the unique region of the PKD1 locus and was recovered from an adult lymphocyte libary. In order to complete the rest of the PKD1 transcript, fourteen new cDNAs were sequenced to completion, four cDNAs were partially sequenced and an additional 20 cDNAs were mapped against   cGGG10.   



  Additional data was obtained from RT-PCR products of the renal cell carcinoma cell line SW839 (ATCC).



   Overlapping partial cDNAs described below were isolated from lymphocyte and fetal kidney libraries. In this way, a 14 kb transcript was assembled starting from the 3' until the
CpG island was reached. It is assumed that the   5'end    of the
PKD1 trancript has been located. No other clones further upstream were recovered upon further screening those cDNA  libraries that had provided the majority of the cDNAs which were used to assemble the full length PKD1 cDNA.



   The cDNAs FK7 and FK11 were recovered from a fetal (gestation age of 14-16 weeks) kidney cDNA library using KG8 cDNA as a probe. This library was constructed with the
Superscript Lambda System from (Gibco/BRL), using oligo d(T) primed cDNA. FK7 and   FKl1    were recovered as SAlI inserts.



  The cDNAs designated BK156, BK194, UN49 and UN52 were recovered from a lymphocyte cell library and pulled by using
FK7 as a probe. UN34 was recovered from the same library by hybridizing with a ScaI-SaII   5'end    probe of FK7. UN53, UN54 and UN59 were recovered from the same lymphocyte library (M.



  Owen laboratory, ICRF; Dunne, PhD thesis, 1994) by double screening clones that were both negative when screening with an FK7 probe and positive when screening with BK156 and UN52.



  The cDNA   NKG11    was recovered from a lymphocyte library screened with cGGG10 and was described previously (Germino,
G.G., Weinstat-Saslow, D., Himmelbauer, H., Gillespie G.A.J.,
Somlo, S., Wirth, B., Barton, N., Harris, K.L., Frischauf,   A.M.    and Reeders,   S.T.    (1992) Genomics, 13:144-151). ). The cDNA named Fhkb21 was obtained from a Clonetech fetal kidney library using BK156 as a probe. MSK3 was obtained by probing an adult kidney library (Clonetech) with a probe from   5'end    of KG8. MSK4 was obtained by nested RT-PCR from primers spanning from exons 7-8 to exons 13-14, followed by second round of PCR with internal primers in exon 8 and exon 13.



   11.1.3. cDNA   SEQUENCING   
 The cDNAs were sequenced to 5-fold coverage by primer walking and/or subloning small fragments into M13 or pBluescript. All cDNA sequences were compared to the cGGG10 cosmid sequence to assess whether they were from the correct locus and to determine intron/exon boundaries. Discrepancies were resequenced to determine whether the differences were genuine. Some of the cDNAs described above were clearly different from the genomic sequence, suggesting that these cDNAs were encoded by another locus.  



   MSK3, FK7 and   FK1l    were obtained using a PKD1-specific probe   (KG8)    were found to be 100% identical to genomic sequence. The cDNA and UN49, which showed 99% identity, is possibly PKD1-specific. BK241, BK194, UN52, UN53, UN54 and
UN59, BK156,   Fhkb21    and   NKG11    were   96-98k    homologous to the cGGG10 defined exon sequence, and thus were assumed to have originated fromt the duplicated loci. In general, differences between genomic cDNA were nucleotide differences scattered through out the cDNA sequence. One exception is
BK194, which has an extra CAG at position 1863 of the previously published partial sequence and arose from alternative splicing of exon 33.

  Another exception is BK241 that has an insertion of the following sequence in a tandem repeat of TTATCAATACTCTGGCTGACCATCGTCA at position 1840 of the previously published sequence (European PKD1 Consortium).



  This sequence was not included in the authentic, full-length
PKD1 cDNA because it arose from the duplicated loci would produce a frame shift in the ccoding region of the PKD1 transcript. Except for BK241, cDNAs in the UN and BK series that overlap with each other are more identical to themselves than to the genomic sequence.



   All sequence assembly was performed using the Staden package XBAP (Dear, S. and Staden R. (1991). Nucleic Acid
Res. 19:3907-3911.)
   11.1.4.    PROTEIN HOMOLOGY SEARCHES
 The PKD1 derived amino acid sequence was subjected to various sequence analysis methods   (Koonin,    E.V., Bork, P. and
Sanders, C. (1994) Yeast chromosome III: new gene functions.



  EMBO 13:493-503). For identifying homologues, initial (SWISSPROT, PIR, GENPEPT, TREMBL, EMBL, GENBANK, NRDB) database searches were performed using the blast series of programs (Altschul, S.F. and Lipman, D.J., 1990, Proc. Natl.



  Acad. Sci. USA 87:5509-5513) by applying filter for compositionally biased regions. (Altschul, S.F. et al., 1994, Nat. Genet. 6:119-129). By default, the BLOSUM62 amino acid exchange matrix was used   (Henikoff,    S. and Henikoff J.G.  



  (1993). Proteins 17:97-61). In order to reveal additional candidate preoteins that might be homologous to PKD1, the
BLOSUM45 and PAM240 matrices were also applied. Putative homolgoues with a blast p-value below 0.1 were studied in detail. Multiple alignments of the candidate domains were carried out using CLUSTALW (Thompson, J.D., Higgins, D.G. and
Gibson, T. (1994). Nucleic Acid Res. 22:4673-4680) and pattern (Rohde, K. and Bork, P. (1993). Comput. Appl.



  Biosci. 9:183-189), motifs and profiles (Grisbskov, M.,
McLachlan, A.D. and Eisenberg, D. (1987) Proc. Natl. Acad.



  Sci. USA 84:4355-4358 were derived. With all these constructs interactive database searches were performed.



  Results of these database searches were used for improving the multiple alignments that were then used for the next round of database searches. The final multiple alignment containing all retrieved members of a module family was then used as input for the secondary structure predictions (Rost,
B. and Sander, C. (1994). Proteins 19:55-872).



   11.1.5. SSCP ANALYSIS
 Single-Stranded Conformational Analysis (SSCP) was performed as follows: 50ng of total genomic DNA was amplified by PCR. In addition to the genomic DNA, each PCR reaction contained 1 picomole of each primer (see below), 0.1   p1      32P    dATP (Amersham), 0.2   41    in AmpliTaq (Pharmacia), in PCR buffer with a final Mg2+ of 1.5 mM in a final volume of 20   41.   

 

  The amplification was performed for 25 cycles, each consisting of   94"    C. for 30 seconds,   60     C. for 30 seconds, and 720 for 60 seconds.



   Intronic primers F25 and Mill-lR were utilized for the initial SSCP evaluation. The fragment amplified with these primers overlaps with the 5' end of KG8. Subsequently, the primers F31 and R35 were ussed to amplify the fragment used to sequence the PKD1 mutation.  



   Primers: F25 (5' TCGGGGCAGCCTCTTCCTG 3');
 Mill-lR (5' TACAGGGAGGGGCTAGGG 3');
 F31 (5' TGCAACTGCCTCCTGGAGG 3')
 R35 (5' GGTCTGTCTCTGCTTCCC 3')
 One microliter of each sample was diluted into loading dye (95% formamide, 20 mM NaOH, 1 mM EDTA, xylene cyanol, bromophenol blue) denatured at 980C for 5 minutes, cooled on ice and loaded onto a 10% (50:1 acrylamide:bisacrylamide) polyacrylamide gel containing 10% glycerol. The gel was run at 40C., 50 watts, for 3 hours. Exposure was overnight on phosphoimager plates.



   Amplified DNA from the one individual with a variant pattern was then reamplified using KG8-F31 and KG8-R35 primers and the above-described PCR conditions. Both reamplified strands were then sequenced using standard procedures for cycle sequencing of PCR products. 32P-dCTP incorporation was used.



   11.2 RESULTS
 A series of overlapping cosmid clones spanning the predicted PKD1 region has been described (Germino, G.G.,
Weinstat-Saslow, D., Himmelbauer, H., Gillespie G.A.J.,
Somlo, S., Wirth, B., Barton, N., Harris, K.L., Frischauf,
A.M. and Reeders, S.T. (1992). Genomics, 13:144-151). The integrity of the cosmid contig was confirmed by long-range restriction mapping and genetic linkage analysis of polymorphic sequences derived from the cosmids. Three cosmids   (cGGG1,    cGGG10 and   cDEB11,    from centromere to telomere) form a contig that includes the 3' end of the adjacent gene, TSC2, (cDEB11) and spans over 80 kilobases centromeric.: At the proximal end of cGGglO, there is a CpG island represented by the Not I site, N54T (FIG.   1A).   



   In order to identify transcripts from the region, the cosmid clones were hybridized to a set of five cDNA libraries. KG8, a cDNA corresponding to the distal 3.2kb of the PKD1 sequence (which is located on cosmid cDEB11), was mapped using a panel of somatic cell hybrids, and found to hybridize to a single locus on chromosome 16p13. Sequence  analysis confirmed that KG8 contains the polyadenylated 3'end of a gene and has an open reading frame (ORF) of 2100 bp and a 1068 bp 3' untranslated region. KG8 was also found to contain a polymorphic (CA) microsatellite repeat. Analysis of this repeat in a large number of PKD1 kindreds revealed no recombination   (supra.)   
 To obtain clones extending 5' of KG8, the cosmids   cGGGlO    and cDEB11 were hybridized to different cDNA libraries.

  When some of the positive clones obtained from these screens were analyzed using somatic cell hybrid panels, they were found to hybridize strongly to several loci on chromosome 16 in addition to the PKD1 region. The restriction maps of the hybridizing loci were so similar that it was concluded that a series of recent duplications of part of the PKD1 gene had occurred (excluding the PKD1 region from which the KG8 cDNA is derived) which had given rise to several PKD1-like genomic segments. This sequence duplication had been reported by the
European PKD1 Consortium (EPKDC, 1994, Cell 77:881-894).



   Preliminary sequence analysis of the cDNA clones revealed that the PKD1 and PKD1-like loci give rise to two or more transcripts sharing   95-98k    sequence identity. Because of the high degree of similarity between PKD1 and PKD1-like transcripts, therefore, it was not possible to determine the correct full-length PKD1 cDNA sequence by simply assembling overlapping partial cDNA clones.



   To begin to determine the sequence of the authentic PKD1 transcript, therefore, it was concluded that genomic PKD1 sequence should be compared to that of the PKD1 specific and
PKDl-like cDNAs homologous to the genomic sequence. To that end, the entire cGGG10 cosmid and PKD1 exon-containing parts of the cDEB11 cosmid were sequenced, as described below.



     11.2.1 SEQUENCE    OF THE GENOMIC REGION OF THE PKD1 LOCUS
 The duplicated portion of the PKD1 gene is largely contained within the cosmid cGGG10. Prior to sequencing cGGG10, the integrity of the clone was established in several ways. First, the restriction map of cGGG10 was compared with  map of the genomic DNA from the PKD1 region. Second, restriction maps of the overlapping portions of cGGG1 and cDEB11 were compared with cGGG10. Third, sequences derived from cGGG10 and overlapping portions of cDEB11 showed 100% similarity. Finally, a P1 phage, PKD1521, was obtained by screening a genomic   P1    library with primers from the TSC2 gene, which maps near the PKD1 gene. No sequence differences were obtained between PKD 1521 and   cGGG10.   



   It was necessary to pursue several approaches to obtain the sequence of cGGG10 (see Section 11.1, above). Briefly, due to the difficulty of sequence certain regions, modifications to standard automated sequencing chemistries had to be made. Both dye terminator and dye primer sequence was used, when appropriate, with several different regions.



  Further, different polymerases and different meltng and polymerization conditions were necessary to optimize the quality of the nucleotide sequence. When sequencing across the CpG island at the 5' end of the PKD1 gene, in addition to modifying the polymerization step, single-stranded templates were used.



   A final ten fold redundancy was achieved for the cGGGlO cosmid in order to be able to accurately compare the genomic sequence with that of the PKD1 specific and PKD1-like cDNAs homologous to this cosmid. The cGGG10 sequences were assembled into three contigs of 8 kb, 23 kb and 4.4 kb, separated by 1 kb and 2.2 kb gaps. A two-fold redundancy was obtained for the cDEB11 cosmid, whose sequence was compared to PKD1 locus specific cDNAs in order to obtain intron/exon boundaries of the unique 3'end of the PKD1 gene.



   11.2.2. PKD1 and PKD1-LIKE cDNAs
 In order to identify putative coding regions and intron/exon boundaries, genomic and cDNA sequences were compared. cDNA clones had been identified in two ways.



  First, fragments of cosmids   cGGGlO    and cDEB were hybridized to five cDNA libraries. Second, each cDNA clone was hybridized to fetal kidney and lymphocyte cDNA libraries to  obtain overlapping clones with which to extend the sequence (FIG. 1B).



   When the sequences of overlapping cDNAs were assembled, a PKD1 trancript length of 14.2 kb was obtained. The predominant transcript detected by Northern analysis using the unique sequence KG8 probe is approximately 14 kb, suggesting that the cDNA clones represent the full-length of the PKD1 trancript.



   Restriction and sequence analyses indicate that a CpG island overlaps the 5' end of the sequence. CpG islands hae been found to mark the 5' ends of many genes. Further, the most 5' cDNA clones (UN53, UN54 and UN59) each have identical 5' ends, providing additional evidence that no upstream PKD1 exons were missed (see Section 11.1, above).



   The multiple cDNAs used to assemble the PKD1 trancript along with the genomic sequence are shown in FIGS. 1A and 1B.



  By comaring the sequences of overlapping cDNAs and analyzing the degree of homology between the different cDNAs and genomic sequence, it was possible to distinguish cDNAs encoded by the authentic PKD1 locus frm those encoded y the homologous loci (see Section 11.1, above). The full length
PKD1 trancript constructed from these exons produces a large continuous open reading frame of 12,902 bp.

 

   Significant sequence heterogeneity ws observed in these cDNAs, suggesting that some level of alternative splicing of the primary PKD1 transcript occurs. For this reason, it was sought to isolate a minimum of two cDNAs containing each exon, in order to increase the probability that all exons contributing to the PKD1 transcript were detected. Formally, however, it remains possible that there exist PKD1 transcripts which ccontain exons that are not present in the cDNA clones samples here.



   Exon 17 was found in two cDNA clones (UN34 and BK156) and in the cosmid sequence, but the exon was not incorporated into the final PKD1 transcript. This is due to a number of reasons. First, the cDNA clones in which this exon is found differed from the cosmid and are likely to represent PKD1  like genes, rather than the authentic PKD1 gene (see Section 11.1, above). Second, this exon is not found in FK1, a cDNA which was cloned using a PKD1-specific probe   (KG8).    Finally, when included in the full-length cDNA, this exon introduces a stop codon (743 nucleotides downstream of exon 17) that would producce a truncated protein of 2651 amino acid residues.



  Further studies are needed to assess whether this exon may be used in diffferent splice combinations in locus-specific trancripts. An ADPKD patient with a heterozygous mutation which introduces a stop codon at position 10,601 of the PKD1 open reading frame. Other mutations tha truncate the PKD1 protein have also been reported by the European PKDl
Consortium. Therefore, it is unlikely that transcripts which include exon 17 are predomiant forms in the kidney.



   11.2.3.   SEOUENCE    ANALYSIS OF THE PREDICTED PKD1 PROTEIN
 The assembly of 46 PKD1 exons yields a predicted transcript is 14.2 kb in length with 228 bp nucleotides of putative 5' untranslated and 790 nucleotides of 3' untranslated sequence. The authentic PKD1 transcript differs from the reported 3' PKD1 sequence (EPKDC, 1994, Cell 77:881894) due to the presence of two extra cytosines at position 12873 of the PKD1 open reading frame (corresponding to PBP position 4563). This frameshift yielded an erroneous carboxy
PKD1 derived amino acid sequence which contained almost 80 additional amino acid residues. The presence of the two extra cytosines as confirmed with the cosmid sequence derived from   cDEB11.   



   The PKD1 protein derived from the assembled PKD1 transcript is 4304 amino acids in length, with a predicted molecular weight of 462 kilodaltons. The nucleotide sequence encompassing the Met-l codon is CTAACGATGC, which represents an uncommon translation start site (Kozak, M. (1984).



  Nucleic Acids Res. 12:857-872). This methionine was determined to be the putative PKD1 translation start site because it is preceded by an in-frame stop codon 63 bases upstream. Furthermore, the PKD1 coding region begins with a  23 amino acid region which exhibits many of the properties of a signal peptide and corresponding cleavage site (von Hejne,
G. (1986). Nucleic Acids Res. 14:4683-4690. Welling, L.W.



  Grantham, J.J. (1972). J. Clin. Invest. 51:1063-1075).



   In addition to the signal sequence, the identification of five domains that have been identified in other proteins and a newly discovered domain strongly suggests the extracellular location of at least the N-terminal half   o;    the protein. Immediately downstream of the signal sequence there are two leucine-rich repeats (LRRs) (Figure 7A-7B). These
LRRs are flanked on both sides by a cysteine rich regions which have homology to the flanking regions of a subset of other LRRS. LRRs occur in numerous proteins and have been shown to be involved in diverse forms of protein-protein interactions. The number of LRR within the respective proteins varies between 2 and 29 (Kobe B. and Deisenhofer J., 1994, Trends. Biochem. Sci. 19:415-421). Adhesive platelet glycoproteins form the largest group in the LRR superfamily (Kobe B. and Deisenhofer J., 1994, Trends. Biochem.

  Sci.



  19:415-421). The structure of the array of 15 LRRs in porcine ribonuclease inhibitor (RI) has recently been crystallized (Kobe B. and Deisenhofer J., 1995, Nature 374:183-186); the LRRs of the RI protein form a horseshoelike structure that surrounds RNase A (Kobe B. and
Deisenhofer J., 1995, Nature 374:183-186). It has been suggested that proteins containing only a few LRR, like the
PKD1 protein, interact with other proteins via the LRRs in order to form the horseshoe-like superstructure for proteinbinding (Kobe B. and Deisenhofer J., 1994, supra..



   Although LRRs occur in various locations in different proteins, the additional flanking cysteine-rich disulfide bridge-containing domains, define a subgroup of extracellular proteins (Kobe B. and Deisenhofer J., 1994, supra. Only a few proteins have been sequenced so far that contain both, the distinct N-terminal and C-terminal flanking cysteine-rich domains (Figures 7A-7B and 8). Among this group are toll, slit, trk, trkB and trkC, which are all involved in cellular  signal transduction. For example, the Drosophila toll protein is suspected to be involved in either adhesion or signaling required to mediate developmental events such as dorsal-ventral patterning (Hashimoto, C., Hudson, K.L., and
Anderson, K.V., 1988, Cell 52:269-279).

  The Drosophila slit protein is thought to possible mediate interactions between growing axons and the surrounding matrix (Rothberg, J.M.,
Jacobs, J.R., Goodman, C.S., and Artavanis-Tsakonas, S., 1990, Genes and Dev. 4:2169-2187). In vertebrates, these domains are found in the trk family of tyrosine kinase receptors; these proteins may relay cell or matrix adhesive events to the cytoplasm via a small carboxy terminal kinase domain (Schneider, R., Schweider, M., 1991, Oncogene 6:180711). it is interesting to note that all of the proteins with these cysteine-rich domains are involved in extracellular function, many of which relate to cell adhesion. For example, the platelet glycoproteins I and V help mediate the adhesion of platelets to sites of vascular injury. The 5T4 oncofetal trophoblast glycoprotein appears to be highly expressed in metastatic tumors.



   The PKD1 protein also contains a single domain with homologies to C-type (calcium-dependent) lectin proteins (Figures 7A-7B and 8). These domains are believed to be involved in the extracellular binding of carbohydrate residues for diverse purposes, including internalization of glycosylated-enzyme (asialoglycoprotein receptors), participation in extracellular matrix (versican) and cell adhesion (selectins). The classification of C-type lectins has been based on exon organization and the nature and arrangement of domains within the protein. For example, class I (extracellular proteoglycans) and class II (type II transmembrane receptors) all have three exons encoding for the carbohydrate recognition domain (CRD); where as in classes III (collectins) and IV (LEC-CAMS) the domains are encoded by a single exon. 

  The CRD in PKD1 C-type lectin domain does not fit into the above classification because it has a novel combination of protein domains and because it is  encoded by two exons (exons 5 and 6, Figure 6A-6P). Previous analysis has failed to establish a correlation between the type of carbohydrate bound to each C-type lectin and the primary structure of its CRD.



   Exon 10 encodes a LDL-A module (from amino acids 642672, Figures 7A-7B), a cysteine-rich domain of about 40 amino acids in length. This module was originally identified in the LDL-receptor but it is also present extracellular portions of many other proteins, often in tandem arrays (Figure 7A-7B). Because of their hydrophobic nature, these domains have been implicated as ligand-binding regions in LDL receptor-related protein. Other proteins, like the PKD1 protein, that contain a single or nontandem LDL-A, include the complement proteins (DiScipio, R.G., Gehring, M.R.,
Podack, E.R., Kan, C.C. Hugli, T.E., and Fey., G.H., 1984,
Proc. Natl. Acad. Sci. USA 81:7298-7302), calf enterokinase (Kitamoto, Y., Yan, X.W., McCourt, D.W. and Sadler, J.E., 1994, Proc. Natl. Acad. Sci. USA 91:7588-7592) and a sarcoma virus adhesion protein.



   In addition to extracellular protein modules that have been recognized previously, the PKD1 protein a novel domain of approximately 70 amino acids in length, present in 14 copies (Figures 7A-7B and 8). The first one is encoded by exon 5 between the LRRs and the C-type lectin module. The other PKD domains are consecutively placed starting at amino acid 1100 and ending at amino acid 2331 and contained in exons 13, 14, and 15. Profile and motif searches (see
Section 11.1, above) identified several other extracellular proteins that also contain one or more copes of this novel domain, which we call the PKD domain. Whereas all known extracellular modules seem to be restricted to higher organisms, and the few exceptions seem to be evolutionary accidents, it was found that the PKD domain in extracellular parts of proteins from animals, eubacteria and archeabacteria.

  The animal proteins containing an individual
PKD domain are heavily glycosylated, melanoma-associated cell surface proteins, such as melanocyte-specific human pmell7   (Kwon BS., 1993, J. Invest. Derm. (Supplement) 100:134-140), the MMP 115 protein (Mochii, M., Agata, K. and Eguchi, G., 1991, Pigment Cell Res. 4:41-47), and the nmb protein   (Weterman,    M.A.J., Ajubi, N., van Dinter, I. Degen, W., van
Muijen, G., Ruiter D.J. and Bloemers, H.P.J., 1995, Int. J.



  Cancer 60:73-81). The physiological functions of these glycoproteins remains to be elucidated. Four enbacterial extracellular enzymers, three distinct collagenases and lysine-specific achromobacter protease I (API) also contain a single copy of the domain adjacent to their catalytic domains. Curiously, the highest degree of similarity between the collagenases is in the PKD domain. This may suggest that the domain in eukaryotic cells is involved in binding to collagenous domains. Four copies of the PKD domain are also present in the surface layer protein (SlpB) from methanothermus. The SlpB protein is (as is the PMEL17 family) heavily glycosylated and is predicted to be a glycoprotein component of the surface layer.



   The PKD domain is predicted to be a globular domain that contains an antiparallel P-sheet. Although the PKD domains do not contain conserved cysteines, we believe they are extracellular domains because: 1) all identified homologues are extracellular or the PKD domain is in the extracellular part; 2) the first domain (amino add 281-353) is located between other known extracellular modules; and 3) there are no predicted transmembrane regions between the other identified (extracellular) modules and the 13 remaining FKD domains. Whereas the PKD domains in SlpB are very similar, pointing to rather recent duplication; the 14 domains in PKD1 are rather divergent. Even the most conserved (WDFGDG) motif (Fig. 7A-7B) is considerably modified in some of the PKD domains.

  Therefore, it is unlikely that unequal recombination between genomic sequences for motifs is a common source of mutations in this disease.



   Although, it was not possible to identify specific domains in the C-terminal half of the protein, a long region was found which contained similarity to a putative C. elegans  
Chromosome III protein (accession number   Z48544).    A hydrophobic stretch of 60 amino acids from 3986 to 4045 might represent a possible transmembrane domain, but without any clear resemblance to other such domains.



   11.2.4. IDENTIFICATION OF AN
 ADPKD- CAUSING MUTATION
 SSCP analysis was performed on samples obtained from 60 patients, as described, above, in Section 10.1. One variant
ADPKD individual was identified via SSCP. Upon reamplification of amplified DNA from this individual (see
Section 10.1, above), it was revealed that the patient contained a C to T transition at base pair 10,601 (exon 32) of the full-length PKD1 transcript. This mutation created a stop codon (TAG) at PKD1 amino acid position 765 which previously coded for a glutamine (CAG), thus truncating the final   728    amino acid residues which are normally present at the carboxy end of the PKD1 protein and yielding a final mutant protein of 3576 amino acids.

  The mutation was also predicted to create a novel Sty-1 site (CCCTAG); genomic DNA spanning this exon was amplified as before from the patient, his parents, and over 60 other unrelated individuals (120   alleles) .    After Sty-1 digestion, only the patient ZC   (#118)    was heterozygous for an enzyme site. The absence of the sequence change in over 120 alleses establishes this is not a polymorphic variation. The absence of the site in either parent establishes this as a new mutation, which corelates with the appearance of disease. Finally, the predicted impact on the protein (truncation) by itself is highly suggestive that it would impair or alter its function.

  This evidence, even in the absence of examination of the remainer of the gene or transcript in this patient, would be considered generally to be sufficient proof that this mutation is the cause of the disease.  

 

   12. DEPOSIT OF MICROORGANISMS
 The following microorganisms were deposited with the
American Type Culture Collection, Rockville, Maryland on May 27, 1994 and assigned the indicated accession numbers:
 Microorganism ATCC Accession No.



   KG8 69636
 cGGG10 69634
 cDEB11 69635
 The present invention is not to be limited in scope by the specific embodiments described which are intended as single illustrations of individual aspects of the invention, and functionally equivalent methods and components are within the scope of the invention. Indeed, various modifications of the invention, in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.  
EMI94.1     


<tb>



   <SEP> MICROORGANISMS
<tb> Optional <SEP> Sheet <SEP> in <SEP> connection <SEP> with <SEP> the <SEP> microorganism <SEP> referred <SEP> to <SEP> on <SEP> page <SEP> 93. <SEP> lines <SEP> 1-20 <SEP> of <SEP> the <SEP> description'
<tb> A. <SEP> IDENTIFICATION <SEP> OF <SEP> DEPOSIT
<tb>  <SEP> Further <SEP> deposits <SEP> are <SEP> identified <SEP> on <SEP> an <SEP> additional <SEP> sheet
<tb> Name <SEP> of <SEP> depositary <SEP> 'ristiltition
<tb> American <SEP> Type <SEP> Culture <SEP> CoUection
<tb> Address <SEP> of <SEP> depositary <SEP> Instltutlon <SEP> (including <SEP> postal <SEP> code <SEP> and <SEP> country)
<tb> 12301 <SEP> Parklawn <SEP> Drive
<tb> Rockville. <SEP> MD <SEP> 20852
<tb> US
<tb> Date <SEP> of <SEP> deposit' <SEP> May <SEP> 27, <SEP> 1994 <SEP> Accession <SEP> Number <SEP> 69636
<tb> B.

  <SEP> ADDITIONAL <SEP> INDICATIONS <SEP> iin,,r <SEP> blank <SEP> If <SEP> m <SEP> IPPllclb*) <SEP> Thss <SEP> mfrmutHM <SEP> o <SEP> crmlmrd <SEP> n <SEP> mmu <SEP> on <SEP> anacbad <SEP> Sheci
<tb> C. <SEP> DESIGNATED <SEP> STATES <SEP> FOR <SEP> WHICH <SEP> INDICATIONS <SEP> ARE <SEP> MADE'cur,uur
<tb> D. <SEP> SEPARATE <SEP> FURNISHING <SEP> OF <SEP> INDICATIONS <SEP> ' <SEP> (i,s5 <SEP> blsak <SEP> if-i <SEP> if <SEP> st <SEP> laaik)
<tb> The <SEP> mecalion, <SEP> baled <SEP> below <SEP> wi <SEP> b. <SEP> nitt <SEP> Ï <SEP> So <SEP> the <SEP> btNtioNi <SEP> Bw..u <SEP> Sara,' <SEP> lspae"r <SEP> the <SEP> g.n"rai <SEP> natura <SEP> of <SEP> She <SEP> iicm <SEP> e.g..
<tb>

 

  'Accunm <SEP> Nunbu <SEP> ot <SEP> Dsw
<tb>  <SEP> tLea <SEP> wu <SEP> rcccivcd <SEP> with <SEP> the <SEP> Intcmuioarl <SEP> rppliation <SEP> whm <SEP> filed <SEP> (to <SEP> k <SEP> chcdred <SEP> by <SEP> he <SEP> recciviy <SEP> Office)
<tb>  <SEP> O251995 <SEP> iOqI
<tb>  <SEP> hs&num;JQBgk&num;Dn
<tb>  <SEP> O <SEP> The <SEP> dale <SEP> of <SEP> i capt <SEP> (from <SEP> the <SEP> applicant) <SEP> by <SEP> tbe <SEP> IleisuloissI <SEP> Burau <SEP> "
<tb>  <SEP> -as <SEP> ¯¯¯¯¯¯¯¯¯¯¯¯¯¯
<tb>  <SEP> (Ai:"lierizei( <SEP> Ofiiccr)
<tb>   
American Type Culture Collection 

Claims

WHAT IS CLAIMED IS: 1. An isolated nucleic acid containing a nucleotide sequence which encodes a polycystic kidney disease (PKD1) gene product.
2. The isolated nucleic acid of Claim 1 which encodes the amino acid sequence (SEQ ID NO: 2) of the PKD1 gene product depicted in FIG. 6.
3. The isolated nucleic acid of Claim 1 wherein the nucleotide sequence is the nucleotide sequence (SEQ ID NO: 1) depicted in FIg. 6.
4. The isolated nucleic acid Claim 1 which hybridizes under stringent conditions to the complement of the coding sequence of the nucleotide sequence depicted in FIG. 6 (SEQ ID NO: 1), or which hybridizes under less stringent conditions and encodes a functionally equivalent PKD1 gene product.
5. A nucleic acid vector containing the nucleotide sequence of Claim 1, 2, 3 or 4.
6. An expression vector containing the nucleotide sequence of Claim 1, 2, 3 or 4 in operative association with a nucleotide regulatory element that controls expression of the nucleotide sequence in a host cell.
7. An antisense molecule containing the nucleotide sequence of Claim 4.
8. A ribozyme molecule containing the nucleotide sequence of Claim 4.
9. A triple helix molecule containing the nucleotide sequence of Claim 4.
10. The nucleotide vector of Claim 5 which is a plasmid vector.
11. The nucleotide vector of Claim 5 which is a viral vector.
12. A genetically engineered host cell containing the nucleotide sequence of Claim 1, 2, 3 or 4.
13. A genetically engineered host cell containing the nucleotide sequence of Claim 1, 2, 3 or 4 in operative association with a regulatory element that controls expression of the nucleotide sequence in the host cell.
14. A substantially pure PKD1 gene product.
15. The substantially pure PKD1 gene product of Claim 14 wherein the gene product contains the amino acid sequence (SEQ ID NO: 2) depicted in FIG. 6.
15. An antibody that immunospecifically binds to a PKD1 gene product.
16. A method for diagnosing autosomal dominant polycystic kidney disease, comprising detecting a mutant PKD1 gene or gene product in a patient sample.
17. A method for treating autosomal dominant polycystic kidney disease, comprising administering an effective amount of a compound to a patient in need of such treatment, which compound inhibits the synthesis, expression or activity of a mutant PKD1 gene product.
18. The method of Claim 17 in which the compound is an antisense or ribozyme molecule that blocks translation of mutant PKD1 mRNA.
19. The method of Claim 18 in which the compound is a nucleotide that is complementary to the 5' region of the PKD1 gene, and blocks transcription of the PKD1 gene via triple helix formation.
20. The method of Claim 19 further comprising replacing the mutant PKD1 gene with a normal allele, or replacing the mutant PKD1 gene product with a normal PKD1 gene product.
21. The method of Claim 19 in which the compound is an antibody that immunospecifically binds and inactivates the mutant PKD1 gene product.
22. A method for treating autosomal dominant polycystic kidney disease, comprising administering a normal allele of the PKD1 gene to a patient in need of such treatment, so that the normal PKD1 allele is expressed in the patient.
23. A method for treating autosomal dominant polycystic kidney disease, comprising administering an effective amount of a normal PKD1 gene product to a patient in need of such therapy.
24. A method of measuring the presence of a PKD1 gene product in a sample, comprising: (a) contacting the sample suspected of containing a PKD1 gene product with an antibody that binds to the PKD1 gene product under conditions which allow for the formation of reaction complexes comprising the antibody and the PKD1 gene product; (b) detecting the formation of reaction complexes comprising the antibody and PKD1 gene product in the sample, in which detection of the formation of reaction complexes indicates the presence of the PKD1 gene product in the sample.
25. The method of Claim 24 in which the antibody is bound to a solid phase support.
26. The method of Claim 24 in which the PKD1 gene product is bound to a solid phase support.
27. The method of Claim 25 or 26 which additionally comprises contacting the sample with a labeled PKD1 gene product in step (a), and removing unbound substances prior to step (b), in which a decrease in the amount of reaction complexes comprising the antibody and the labelled PKD1 gene product indicates the presence of the PKD1 gene product in the sample.
28. A method of evaluating the level of PKD1 gene product in a biological sample comprising: (a) detecting the formation of reaction complexes in a biological sample according to the method of Claim 24; and (b) evaluating the amount of reaction complexes formed, which amount of reaction complexes corresponds to the level of PKDl gene product in the biological sample.
29. A method of detecting or diagnosing the presence of a disease associated with elevated or decreased levels of PKD1 gene product in a mammalian subject comprising: (a) evaluating the level of PKD1 gene product in a biological sample from mammalian subject according to Claim 28; and (b) comparing the level detected in step (a) to a level of PKD1 gene product present in normal subjects or in the subject at an earlier time, in which an increase or a decrease in the level of the PKD1 gene product as compared to normal levels indicates a disease condition.
30. A method for monitoring a therapeutic treatment of a disease associated with elevated or decreased levels of PKD1 gene product in a mammalian subject, comprising evaluating the levels of the PKD1 gene product in a series of biological samples obtained at different time points from a mammalian subject undergoing a therapeutic treatment for a disease associated with elevated or decreased levels of PKD1 gene product, according to the method of Claim 28.
31. The method according to Claim 29 or 30 wherein the disease associated with decreased levels of PKD1 gene product is selected from the group consisting of polycystic kidney disease, and acquired cystic disease.
32. A test kit for measuring the presence of or amount of PKD1 gene product in a sample, comprising (a) an antibody that immunospecifically binds to a PKDl gene product; (b) means for detecting binding of the anti-PKD1 gene product antibody to PKD1 gene product in a sample; (c) other reagents; and (d) directions for use of the kit.
33. A pharmaceutical composition for treating polycystic kidney disease in a mammal, comprising the PKD1 gene product of Claim 14 and a pharmaceutically acceptable carrier.
34. A method for treating polycystic kidney disease in a mammal comprising administering an amount of a pharmaceutical composition of Claim 33 effective to ameliorate the symptoms of polycystic kidney disease.
35. A method for treating polycystic kidney disease in a mammal comprising increasing the expression of a protein encoded by the nucleic acid of Claim 1, 2, 3 or 4.
PCT/US1995/007079 1994-06-03 1995-06-02 Identification of polycystic kidney disease gene, diagnostics and treatment WO1995034573A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU27661/95A AU2766195A (en) 1994-06-03 1995-06-02 Identification of polycystic kidney disease gene, diagnostics and treatment

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US25352494A 1994-06-03 1994-06-03
US253,524 1994-06-03
US41358095A 1995-03-30 1995-03-30
US413,580 1995-03-30

Publications (2)

Publication Number Publication Date
WO1995034573A1 true WO1995034573A1 (en) 1995-12-21
WO1995034573A9 WO1995034573A9 (en) 1996-03-14

Family

ID=26943335

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/007079 WO1995034573A1 (en) 1994-06-03 1995-06-02 Identification of polycystic kidney disease gene, diagnostics and treatment

Country Status (3)

Country Link
US (1) US5891628A (en)
AU (1) AU2766195A (en)
WO (1) WO1995034573A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997032024A1 (en) * 1996-03-01 1997-09-04 Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Allele suppression
WO1997039102A2 (en) * 1996-04-18 1997-10-23 Genetics Institute, Inc. Secreted proteins
WO1997039030A2 (en) * 1996-04-18 1997-10-23 Genetics Institute, Inc. Secreted proteins
WO1998024894A2 (en) * 1996-12-06 1998-06-11 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Pkd1 fragments with bonding areas for pkd1-specific antibodies
WO2000024780A2 (en) * 1998-10-26 2000-05-04 Genzyme Corporation Compositions and methods for treating polycystic kidney disease
WO2000040711A2 (en) * 1999-01-06 2000-07-13 California Institute Of Technology Polycystic kidney disease gene homologs required for male mating behavior in nematodes and assays based thereon
WO2001012796A2 (en) * 1999-06-01 2001-02-22 Devgen Nv Pkd constructs and screening methods
US6280739B1 (en) 1996-04-18 2001-08-28 Genetics Institute, Inc. Method of inhibiting angiogenesis using secreted proteins
US6713457B2 (en) 1995-09-21 2004-03-30 Gwenyth Jane Farrar Strategy for suppressing the expression of an endogeneous gene by using compounds that are able to bind to the non-coding regions of the gene to be suppressed
US6723557B1 (en) 1999-01-06 2004-04-20 California Institute Of Technology Caenorhabditis elegans LOV-1 gene
US7138378B1 (en) 1996-04-02 2006-11-21 Optigen Patents Limited Genetic suppression and replacement
US8551970B2 (en) 1996-04-02 2013-10-08 Optigen Patents Limited Genetic suppression and replacement

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995018226A1 (en) * 1993-12-24 1995-07-06 University Of Wales College Of Medicine Tuberous sclerosis 2 gene and uses thereof
EP1105537B1 (en) 1998-08-19 2005-11-16 Bioventures, Inc. Method for determining polynucleotide sequence variations
US20030138834A1 (en) * 1998-08-19 2003-07-24 Dawson Elliott P. Method for determining polynucleotide sequence variations
US20040121394A1 (en) * 1998-08-19 2004-06-24 Dawson Elliott P. Method for determining polynucleotide sequence variations
US6864355B1 (en) 2000-05-02 2005-03-08 Yale University Inhibition of NF-κB activation by blockade of IKKβ-NEMO interactions at the NEMO binding domain
AU8021500A (en) * 1999-10-13 2001-04-23 Johns Hopkins University School Of Medicine, The Methods of diagnosing and treating hepatic cell proliferative disorders
US6709818B1 (en) 1999-10-13 2004-03-23 The Johns Hopkins University School Of Medicine Methods of diagnosing and treating hepatic cell proliferative disorders
BR0015100A (en) * 1999-10-26 2003-07-15 Deltagen Inc Transgenic mice containing trp gene disruptions
US6638726B1 (en) * 2000-01-06 2003-10-28 Mount Sinai School Of Medicine Of New York University Screening methods for compounds useful in the treatment of polycystic kidney disease
DE60232785D1 (en) 2001-03-14 2009-08-13 Myriad Genetics Inc TSG101 GAG INTERACTION AND ITS USE
WO2002074930A2 (en) * 2001-03-20 2002-09-26 The Pennsylvania State Research Foundation Trans inteins for protein domain shuffling and biopolymerization
WO2002083839A2 (en) * 2001-03-30 2002-10-24 Amersham Biosciences Ab P450 single nucleotide polymorphism biochip analysis
US6841352B2 (en) 2001-06-29 2005-01-11 Myriad Genetics, Inc. Mating-based method for detecting protein—protein interaction
EP1428024A2 (en) * 2001-09-21 2004-06-16 Caprion Pharmaceuticals, Inc. Preparation of highly-purified plasma membranes
US6916619B2 (en) * 2001-10-12 2005-07-12 Athena Diagnostics, Inc. Compositions and methods for genetic analysis of polycystic kidney disease
US7273701B2 (en) * 2001-10-12 2007-09-25 Athena Diagnostics, Inc. Compositions and methods for genetic analysis of polycystic kidney disease
WO2007019376A2 (en) * 2005-08-03 2007-02-15 Rq Bioscience, Inc. Methods and compositions for diagnosis of iga-and igm-mediated kidney diseases
AU2006308863A1 (en) * 2005-10-31 2007-05-10 Janssen Pharmaceutica N.V. A polypeptide complex of TRPM8 and calmodulin and its uses thereof
US20100159450A1 (en) 2006-06-23 2010-06-24 Susanne Wagner Dpyd gene variants and use thereof
EP2097448A4 (en) 2006-12-22 2010-07-21 Univ Utah Res Found Method of detecting ocular diseases and pathologic conditions and treatment of same
US20100144545A1 (en) * 2007-03-23 2010-06-10 Arlene Chapman Arrays, Systems, and Methods of Using Genetic Predictors of Polycystic Diseases
WO2009131957A2 (en) 2008-04-21 2009-10-29 Institute For Oneworld Health Compounds, compositions and methods comprising oxadiazole derivatives
US8236838B2 (en) * 2008-04-21 2012-08-07 Institute For Oneworld Health Compounds, compositions and methods comprising isoxazole derivatives
WO2009131947A2 (en) * 2008-04-21 2009-10-29 Institute For Oneworld Health Compounds, compositions and methods comprising pyridazine derivatives
WO2009131958A2 (en) * 2008-04-21 2009-10-29 Institute For Oneworld Health Compounds, compositions and methods comprising triazine derivatives
WO2010033626A1 (en) * 2008-09-19 2010-03-25 Institute For Oneworld Health Compounds, compositions and methods comprising imidazole and triazole derivatives
US8511216B2 (en) * 2009-03-30 2013-08-20 Kanzaki Kokyukoki Mfg. Co., Ltd. Hydraulic actuator unit
US8343976B2 (en) * 2009-04-20 2013-01-01 Institute For Oneworld Health Compounds, compositions and methods comprising pyrazole derivatives
WO2013019169A1 (en) 2011-08-01 2013-02-07 Institute For Oneworld Health Phosphate prodrugs
KR20140082796A (en) 2011-10-14 2014-07-02 제넨테크, 인크. ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
KR102162324B1 (en) 2015-10-30 2020-10-07 제넨테크, 인크. Anti-HtrA1 antibodies and methods of use thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5654170A (en) * 1994-10-12 1997-08-05 Johns Hopkins University Polycystic kidney disease gene

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
AMERICAN JOURNAL OF HUMAN GENETICS, Volume 46, issued 1990, GERMINO et al., "Identification of A Locus Which Shows No Genetic Recombination with the Autosomal Dominant Polycystic Kidney Disease Gene on Chromosome 16", pages 925-933. *
CLINICAL CHEMISTRY, Volume 35, issued 1989, REEDERS et al., "Mapping the Locus of Autosomal Dominant Polycystic Kidney Disease: Diagnostic Application", pages B13-16. *
GENOMICS, Volume 13, issued 1992, GERMINO et al., "The Gene for Autosomal Dominant Polycystic Kidney Disease Lies in a 750-Kb CpG-Rich Region", pages 144-151. *
GENOMICS, Volume 13, issued 1992, HIMMELBAUER et al., "Human-Mouse Homologies in the Region of the Polycystic Kidney Disease Gene (PKD1)", pages 35-38. *
GENOMICS, Volume 13, issued 1992, SMOLO et al., "Fine Genetic Localization of the Gene for Autosomal Dominant Polycystic Kidney Disease (PKD1) with Respect to Physically Mapped Markers", pages 152-158. *
GENOMICS, Volume 18, issued 1993, KIMBERLING et al., "Autosomal Dominant Polycystic Kidney Disease: Localization of the Second Gene to Chromosome 4q13-q23", pages 467-472. *
HUMAN GENETICS, Volume 90, issued 1993, WRIGHT et al., "A Study of Genetic Linkage Heterogeneity in 35 Adult-Onset Polycystic Kidney Disease Families", pages 569-571. *
JOURNAL OF MEDICAL GENETICS, Volume 27, issued 1990, BREUNING et al., "Mapping of 16 Polymorphic Loci on the Short Arm of Chromosome 16 Close to the Polycystic Kidney Disease Gene (PKD1)", pages 603-613. *
NATURE GENETICS, Volume 1, issued July 1992, REEDERS, "Multilocus Polycystic Disease", pages 235-237. *
NUCLEIC ACIDS RESEARCH, Volume 18, Number 23, issued 1990, GILLESPIE et al., "Cosmid Walking and Chromosome Jumping in the Region of PKD1 Reveal a Locus Duplication and Three CpG Islands", pages 7071-7075. *

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6713457B2 (en) 1995-09-21 2004-03-30 Gwenyth Jane Farrar Strategy for suppressing the expression of an endogeneous gene by using compounds that are able to bind to the non-coding regions of the gene to be suppressed
WO1997032024A1 (en) * 1996-03-01 1997-09-04 Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Allele suppression
US8551970B2 (en) 1996-04-02 2013-10-08 Optigen Patents Limited Genetic suppression and replacement
US7138378B1 (en) 1996-04-02 2006-11-21 Optigen Patents Limited Genetic suppression and replacement
US6280739B1 (en) 1996-04-18 2001-08-28 Genetics Institute, Inc. Method of inhibiting angiogenesis using secreted proteins
WO1997039102A2 (en) * 1996-04-18 1997-10-23 Genetics Institute, Inc. Secreted proteins
WO1997039030A2 (en) * 1996-04-18 1997-10-23 Genetics Institute, Inc. Secreted proteins
WO1997039102A3 (en) * 1996-04-18 1997-11-27 Genetics Inst Secreted proteins
WO1997039030A3 (en) * 1996-04-18 1998-03-26 Genetics Inst Secreted proteins
WO1998024894A2 (en) * 1996-12-06 1998-06-11 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Pkd1 fragments with bonding areas for pkd1-specific antibodies
WO1998024894A3 (en) * 1996-12-06 1998-07-16 Deutsches Krebsforsch Pkd1 fragments with bonding areas for pkd1-specific antibodies
WO2000024780A3 (en) * 1998-10-26 2000-11-09 Genzyme Corp Compositions and methods for treating polycystic kidney disease
JP2002528462A (en) * 1998-10-26 2002-09-03 ジェンザイム・コーポレーション Compositions and methods for treating polycystic kidney disease
AU770054B2 (en) * 1998-10-26 2004-02-12 Genzyme Corporation Compositions and methods for treating polycystic kidney disease
WO2000024780A2 (en) * 1998-10-26 2000-05-04 Genzyme Corporation Compositions and methods for treating polycystic kidney disease
WO2000040711A3 (en) * 1999-01-06 2000-09-21 California Inst Of Techn Polycystic kidney disease gene homologs required for male mating behavior in nematodes and assays based thereon
US6723557B1 (en) 1999-01-06 2004-04-20 California Institute Of Technology Caenorhabditis elegans LOV-1 gene
US6849717B1 (en) 1999-01-06 2005-02-01 California Institute Of Technology Polycystic kidney disease gene homologs required for male mating behavior in nematodes and assays based thereon
WO2000040711A2 (en) * 1999-01-06 2000-07-13 California Institute Of Technology Polycystic kidney disease gene homologs required for male mating behavior in nematodes and assays based thereon
WO2001012796A3 (en) * 1999-06-01 2001-08-23 Titus Kaletta PKD constructs and screening methods
WO2001012796A2 (en) * 1999-06-01 2001-02-22 Devgen Nv Pkd constructs and screening methods

Also Published As

Publication number Publication date
AU2766195A (en) 1996-01-05
US5891628A (en) 1999-04-06

Similar Documents

Publication Publication Date Title
US5891628A (en) Identification of polycystic kidney disease gene, diagnostics and treatment
WO1995034573A9 (en) Identification of polycystic kidney disease gene, diagnostics and treatment
US5817762A (en) Mammalian tub protein
AU749198B2 (en) A novel human G-protein coupled receptor
JP2008167759A (en) Nphs2 gene relating to steroid-resistant nephrotic syndrome and protein encoded by the same, and use for treatment and diagnosis thereof
US5861239A (en) Methods for identifying compounds that modulate mammalian tub protein activity
US20020072101A1 (en) Novel human nucleic acid molecules and polypeptides encoding cation channels
US6987178B2 (en) Human uncoupling proteins and polynucleotides encoding the same
WO2000005373A2 (en) Methods and compositions for the diagnosis and treatment of body weight disorders, including obesity
AU774850B2 (en) Human seven-transmembrane receptors
US6310182B1 (en) Compositions for the diagnosis and treatment of Chediak-Higashi syndrome
WO1997034914A9 (en) Compositions for the diagnosis and treatment of chediak-higashi syndrome
AU777909B2 (en) Mammalian cortexin-like proteins and polynucleotides encoding the same
AU784052B2 (en) Novel human proteins and polynucleotides encoding the same
AU773638B2 (en) MEG-4 protein
US20060019248A1 (en) Mutant sh-3 binding protein compositions and methods
AU775409B2 (en) Meg-3 protein
JP2004531209A (en) Novel human 7TM protein and polynucleotide encoding the same
JP2004506447A (en) Novel human 7TM protein and polynucleotide encoding the same
AU5118399A (en) Methods and compositions for the diagnosis and treatment of body weight disorders, including obesity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AU BB BG BR BY CA CN CZ EE FI GE HU IS JP KE KG KR KZ LK LR LT LV MD MG MN MW MX NO NZ PL RO RU SD SG SI SK TJ TM TT UA UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 1-93,DESCRIPTION,PAGES 94-98,CLAIMS,REPLACED BY NEW PAGES BEARING THE SAME NUMBER,PAGES 1/21-21/21,DRAWINGS,REPLACED BY NEW PAGES 1/28-28/28;DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA